EP2102244A2 - Gegen metalloproteinase aus der adam-familie gerichtete aminosäuresequenzen und diese enthaltende polypeptide zur behandlung von mit adam in zusammenhang stehenden krankheiten und störungen - Google Patents

Gegen metalloproteinase aus der adam-familie gerichtete aminosäuresequenzen und diese enthaltende polypeptide zur behandlung von mit adam in zusammenhang stehenden krankheiten und störungen

Info

Publication number
EP2102244A2
EP2102244A2 EP07857865A EP07857865A EP2102244A2 EP 2102244 A2 EP2102244 A2 EP 2102244A2 EP 07857865 A EP07857865 A EP 07857865A EP 07857865 A EP07857865 A EP 07857865A EP 2102244 A2 EP2102244 A2 EP 2102244A2
Authority
EP
European Patent Office
Prior art keywords
amino acid
acid sequences
acid sequence
seq
sequences
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
EP07857865A
Other languages
English (en)
French (fr)
Inventor
Christophe Blanchetot
Michael John Scott Saunders
Johannes Joseph Wilhelmus De Haard
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ablynx NV
Original Assignee
Ablynx NV
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ablynx NV filed Critical Ablynx NV
Priority to EP12164303A priority Critical patent/EP2514767A1/de
Publication of EP2102244A2 publication Critical patent/EP2102244A2/de
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/22Immunoglobulins specific features characterized by taxonomic origin from camelids, e.g. camel, llama or dromedary
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • AMTNO ACTD SEQUENCES DTRECTED AGATNST A METAT .LOPROTETNASE
  • the present invention relates to amino acid sequences that are directed against (as defined herein) metalloproteinases from the ADAM family, as well as to compounds or constructs, and in particular proteins and polypeptides, that comprise or essentially consist of one or more such amino acid sequences (also referred to herein as ""amino acid sequences of the invention', ""compounds of the invention', and “"polypeptides of the invention', respectively).
  • the invention also relates to nucleic acids encoding such amino acid sequences and polypeptides (also referred to herein as ""nucleic acids of the invention” or “"nucleotide sequences of the invention'); to methods for preparing such amino acid sequences and polypeptides; to host cells expressing or capable of expressing such amino acid sequences or polypeptides; to compositions, and in particular to pharmaceutical compositions, that comprise such amino acid sequences, polypeptides, nucleic acids and/or host cells; and to uses of such amino acid sequences or polypeptides, nucleic acids, host cells and/or compositions, in particular for prophylactic, therapeutic or diagnostic purposes, such as the prophylactic, therapeutic or diagnostic purposes mentioned herein.
  • Other aspects, embodiments, advantages and applications of the invention will become clear from the further description herein.
  • ADAM metalloproteinases disintegrins form a well-known family of proteases (or proteinases) that mediates ectodomain shedding. Many of the processing events that include the release of cytokines, shedding of cell surface molecules, release of growth factors and cleavage of amyloid precursor protein (APP), are all shown to be mediated by metalloproteases.
  • the ADAM (A disintegrin And Metalloproteases) family includes proteins containing a disintegrin-like and metalloprotease-like domain).
  • the ADAMTS contain a thrombospondin (TS) domain in addition to the other common domains (and should be considered included within the term ADAM proteinase as used in the specification and claims).
  • TS thrombospondin
  • SVMPs Snake Venom Metalloproteinases
  • ADAM and ADAM- TS form the reprolysin subfamily of metalloprotease. All family members share a common multi-domain structure minimally containing a Pro domain, a metalloprotease (-like) and a disintegrin-like domain.
  • Some ADAM have additional domains like cystein-rich and a transmembrane and cytoplasmic domains.
  • the substrates of ADAM metalloproteinases are also well-known, and for example include CD23, TNFRl, ILl-RII, CX3CL1, TNF-alpha, TNFRl, TNFRII, and TRANCE, as well as the substrates mentioned in Table 1 of the review by Huovila et al., supra.; and Table 1 in the review by Moss et al (2004), supra.
  • ADAM ADAM
  • ADAMl ADAM2, ADAM3B, ADAM4, ADAM5, ADAM6, ADAM7, ADAM8, ADAM9, ADAMlO, ADAMl 1, ADAM 12, ADAM 13, ADAM 14, ADAM 15, ADAM16, ADAM17, ADAM18, ADAM19, ADAM20, ADAM21, ADAM21,
  • ADAMTS13, ADAMTS14, ADAMTS15, ADAMTS16, ADAMTS17, ADAMTS18, ADAMTS 19, ADAMTS20 Reference is also made to the following websites: http://merops.sanger.ac.uk/ and http://www.uta.fi/%7Eloiika/ADAMs/HADAMs.htm.
  • ADAM metalloproteinases Diseases and disorders in which ADAM metalloproteinases are involved as also well- known in the art.
  • ADAM8 allergy, inflammation (asthma, TRAPS) - ADAM9: hematological malignancies
  • ADAMlO chemotaxis, inflammation (asthma, rhinitis) Alzheimer ADAM17/TACE: inflammation, ILlbeta signaling, sepsis, inflammatory, arthritis, diabetes, HIV cachexia, cancer and TNF/EGF dependent diseases
  • ADAM-TS5 aggrecanase 2 and osteoarthritis
  • arthritis - ADAMl 5 atherosclerosis
  • ADAM33 Asthma
  • ADAM 17 is the major physiological TNFalpha converting enzyme and is therefore essential to TNFalpha signaling. Therefore, inhibition of ADAM 17 will be useful in any disease state where TNF antagonist has been validated, such as arthritis, diabetes, HIV cachexia, sepsis and cancer. Reference is for example made to the review articles cited above and the further references cited herein and in the present specification. Moss et al. (2001) and Moss et al. (2004).
  • ADAM 17 is responsible for the processing and release of TGF alpha and EGF. Indeed ADAM 17-/- mice are embryonic lethal with phenotype reminiscent of the TGFalpha-/- and EGFR-/- mice. "Key functions of ADAMs have emerged in ErbB signalling pathways as being sheddases for multiple ErbB ligands. As the ErbB pathway is a validated target for anti-cancer drugs, the upstream activators of ErbB ligands, their sheddases, now enter the spotlight as new drug targets in the ErbB pathway. ADAMs are involved not only in tumour cell proliferation but also in angiogenesis and metastasis. Therefore, strategies targeting ADAMs might be an important complement to existing anti-ErbB approaches.” (See Blobel, supra).
  • ADAMlO and ADAM 17 have been described as important sheddases for cytokines and their receptors, suggesting that these ADAMs are key modulator of cytokines in vivo.
  • the gene ADAM33 was identified as an asthma susceptibility gene involved in airway remodeling.
  • ADAMlO (and to lesser extend ADAM 17) is an alpha- secretase of the amyloid precursor protein (APP). Because cleavage of APP at the alpha site is believed to preclude cleavage at the beta and gamma site (cleavage sites promoting the APP aggregation leading to Alzheimer disease), ADAMlO can be considered as a protective factor in the etiology of Alzheimer's disease. Activation of ADAMlO might prove useful for the treatment of Alzheimer's disease (see for review Allinson 2003 and ref therein).
  • ADAM 12 cleaves and inactivate IGFBP3, a natural inhibitor of IGFl and 2. Because low level of IGF are associated with osteoarthritis and diabetes, the balance between IGFs and IGFBPs is crucial. Interestingly, mice overexpressing ADAM 12 develop more abdominal and body fat. Also ADAM 12-/- mice have reduced adipocytes and partial defect in myogenesis.
  • ADAM9 is expressed in hematological malignancies and might process the EGFR ligand HB-EGF.
  • ADAM8 is a serological and histochemical marker for lung cancer (Ishikawa N, 2004).
  • ADAM8 may be the CD23 shedding enzyme. Soluble CD23 released from cells during allergic response stimulate IgE production. Specific inhibition of CD23 sheddase activity would be an important treatment of allergic reactions.
  • ADAM8 is (over)expressed in tissues around aseptically loosened total hip implants, which are characterized by chronic foreign body inflammation and peri-implant bone loss. This is compatible with a role for ADAM8 in the formation of foreign body giant cells and osteoclasts.
  • ADAM 19 expression has been associated with clinical and structural deterioration involved in renal disease, and ADAM 19 may have a role in the dysfunctional renal allograft state.
  • ADAM8 and ADAM 19 are associated with invasiveness in human primary brain tumors.
  • Snake venom metalloproteinases play an important role in viperid envenoming (hemorrhage, edema, hypertension, hypovolemia, inflammation and necrosis) (see for example Fox et al., supra).
  • ADAM-TS5 is the major aggrecanase (aggrecan is a major component of the cartilage extracellular matrix) responsible of osteoarthritis and inflammatory arthritis.
  • ADAMTS- 13 deficiency gives rise to thrombotic thrombocytopenic purpura (TTP), a life threatening condition (see for example Levy et al., supra)
  • ADAMs may have an influence on the biological pathways, physiological effects, signaling mechanisms and other biological and physiological activities that their respective substrates (and the ligands of such substrates) are involved in.
  • modulation of ADAMs could also play a part in the prevention and treatment of diseases and disorders in which their substrates are involved.
  • metalloproteinases closely related to the ADAM family are the snake venom metalloproteinases such as, without limitation, Aclpref, Acostatin, Acurhagin, Acutolysin A, Atrolysin, Atrolysin A, Atrolysin B, Atrolysin C, Atrolysin E, Atroxase, BAPl, Berythractivase, Bilitoxin-I, Bothropasin, Brevilysin H6, Catrocollastatin, Contortrostatin, Ecarin, Fibrolase, Flavoridin, Flavostatin, Graminelysin I, H2 -Proteinase, HF3, HRlA, HRlB, HR2A, HT-2, HVl, Jararhagin, Jerdonitin, Kaouthiagin, Lebatase, LHF- II, MT-D, RVV-X, Trigramin, VAPl, VLFX
  • the polypeptides and compositions of the present invention can generally be used to modulate, and in particular inhibit and/or prevent, the interaction between a metalloproteinase from the ADAM family and its substrate (and in particular ADAM-mediated ectodomain- shedding, i.e. the ADAM-mediated release of extracellular domains from the substrate) and thus to modulate, and in particular inhibit or prevent, the signalling that is mediated by a metalloproteinase from the ADAM family and/or by its substrate, to modulate the biological pathways in which a metalloproteinase from the ADAM family and/or its substrate is involved, and/or to modulate the biological mechanisms, responses and effects associated with such signalling or these pathways.
  • the polypeptides and compositions of the present invention can be used for the prevention and treatment (as defined herein) of ADAM-related diseases and disorders.
  • ADAM-related diseases and disorders can be defined as diseases and disorders that can be prevented and/or treated, respectively, by suitably administering to a subject in need thereof (i.e.
  • ADAM-related diseases and disorders having the disease or disorder or at least one symptom thereof and/or at risk of attracting or developing the disease or disorder) of either a polypeptide or composition of the invention (and in particular, of a pharmaceutically active amount thereof) and/or of a known active principle active against a metalloproteinase from the ADAM family (or against one of its substrates, and/or against one of the ligands of such a substrate) or a biological pathway or mechanism in which a metalloproteinase from the ADAM family (and/or one of its substrates) is involved (and in particular, of a pharmaceutically active amount thereof).
  • ADAM-related diseases and disorders will be clear to the skilled person based on the disclosure herein, and for example include the diseases and disorders mentioned herein and in the prior art cited herein.
  • polypeptides and compositions of the present invention can be used for the prevention and treatment of ADAM-related diseases and disorders which are characterized by excessive and/or unwanted signalling mediated by a substrate of a metalloproteinase from the ADAM family (or one of the ligands of such a substrate) or by the pathway(s) in which a metalloproteinase from the ADAM family is involved.
  • ADAM-related diseases and disorders will again be clear to the skilled person based on the disclosure herein.
  • amino acid sequences and polypeptides of the invention can for example be used to prevent and/or to treat all diseases and disorders that are currently being prevented or treated with active principles that can modulate a metalloproteinase from the ADAM family-mediated signalling, such as those mentioned in the prior art cited above. It is also envisaged that the polypeptides of the invention can be used to prevent and/or to treat all diseases and disorders for which treatment with such active principles is currently being developed, has been proposed, or will be proposed or developed in future.
  • polypeptides of the present invention may be used for the prevention and treatment of other diseases and disorders than those for which these known active principles are being used or will be proposed or developed; and/or that the polypeptides of the present invention may provide new methods and regimens for treating the diseases and disorders described herein.
  • amino acid sequences that are directed against (as defined herein) a metalloproteinase from the ADAM family, in particular against a metalloproteinase from the ADAM family from a warmblooded animal, more in particular against a metalloproteinase from the ADAM family from a mammal, and especially against human a metalloproteinase from the ADAM family; and to provide proteins and polypeptides comprising or essentially consisting of at least one such amino acid sequence.
  • a metalloproteinase from the ADAM family such as the diseases, disorders and conditions mentioned herein
  • the invention provides amino acid sequences that are directed against (as defined herein) and/or can specifically bind (as defined herein) to a metalloproteinase from the ADAM family; as well as compounds and constructs, and in particular proteins and polypeptides, that comprise at least one such amino acid sequence.
  • the invention provides amino acid sequences that can bind to a metalloproteinase from the ADAM family with an affinity (suitably measured and/or expressed as a K D -value (actual or apparent), a K A -value (actual or apparent), a k on -rate and/or a k ofr rate, or alternatively as an IC 50 value, as further described herein) that is as defined herein; as well as compounds and constructs, and in particular proteins and polypeptides, that comprise at least one such amino acid sequence.
  • amino acid sequences and polypeptides of the invention are preferably such that they: bind to a metalloproteinase from the ADAM family with a dissociation constant (KD) of 10 ⁇ 5 to 10 ⁇ 12 moles/liter or less, and preferably 10 ⁇ 7 to 10 ⁇ 12 moles/liter or less and more preferably 10 "8 to 10 "12 moles/liter (i.e.
  • KD dissociation constant
  • a monovalent amino acid sequence of the invention is preferably such that it will bind to a metalloproteinase from the ADAM family with an affinity less than 500 nM, preferably less than 200 nM, more preferably less than 10 nM, such as less than 500 pM.
  • an amino acid sequence of the invention will usually contain within its amino acid sequence one or more amino acid residues or one or more stretches of amino acid residues (i.e. with each "stretch” comprising two or amino acid residues that are adjacent to each other or in close proximity to each other, i.e. in the primary or tertiary structure of the amino acid sequence) via which the amino acid sequence of the invention can bind to a metalloproteinase from the ADAM family, which amino acid residues or stretches of amino acid residues thus form the "site" for binding to a metalloproteinase from the ADAM family (also referred to herein as the "antigen binding site").
  • amino acid sequences provided by the invention are preferably in essentially isolated form (as defined herein), or form part of a protein or polypeptide of the invention (as defined herein), which may comprise or essentially consist of one or more amino acid sequences of the invention and which may optionally further comprise one or more further amino acid sequences (all optionally linked via one or more suitable linkers).
  • the one or more amino acid sequences of the invention may be used as a binding unit in such a protein or polypeptide, which may optionally contain one or more further amino acid sequences that can serve as a binding unit (i.e.
  • a metalloproteinase from the ADAM family against one or more other targets than a metalloproteinase from the ADAM family), so as to provide a monovalent, multivalent or multispecific polypeptide of the invention, respectively, all as described herein.
  • a protein or polypeptide may also be in essentially isolated form (as defined herein).
  • amino acid sequences and polypeptides of the invention as such preferably essentially consist of a single amino acid chain that is not linked via disulphide bridges to any other amino acid sequence or chain (but that may or may not contain one or more intramolecular disulphide bridges.
  • Nanobodies - as described herein - may sometimes contain a disulphide bridge between CDR3 and CDRl or FR2).
  • one or more amino acid sequences of the invention may be linked to each other and/or to other amino acid sequences (e.g.
  • an amino acid sequence of the invention (or a compound, construct or polypeptide comprising the same) is intended for administration to a subject (for example for therapeutic and/or diagnostic purposes as described herein), it is preferably either an amino acid sequence that does not occur naturally in said subject; or, when it does occur naturally in said subject, in essentially isolated form (as defined herein).
  • amino acid sequences of the invention are preferably directed against human a metalloproteinase from the ADAM family; whereas for veterinary purposes, the amino acid sequences and polypeptides of the invention are preferably directed against a metalloproteinase from the ADAM family from the species to be treated, or at least cross-reactive with a metalloproteinase from the ADAM family from the species to be treated.
  • an amino acid sequence of the invention may optionally, and in addition to the at least one binding site for binding against a metalloproteinase from the ADAM family, contain one or more further binding sites for binding against other antigens, proteins or targets.
  • the efficacy of the amino acid sequences and polypeptides of the invention, and of compositions comprising the same, can be tested using any suitable in vitro assay, cell -based assay, in vivo assay and/or animal model known per se, or any combination thereof, depending on the specific disease or disorder involved.
  • suitable assays and animal models will be clear to the skilled person, and for example include techniques such as BIACORE and FLIPR; commercially available protease assays (for example the assays for ADAM-(TS) available from R&D Systems.
  • the ES003 substrate can be used); cell-based assays that measure sheddase activity (for example for CD23, EGF or TNF, see Weskamp et at, supra and the references cited therein), as well as the assays and animal models used in the experimental part below and in the prior art cited herein.
  • assays can be used that measure the activity of the substrate of ADAM or the signalling or biological activities in which such a substrate (or its natural ligand) in involved.
  • TNF alpha release cleavage
  • ADAM 17 modulators can be tested in this setup.
  • ADAMlO activity can be quantified using soluble CD23 (see for example Weskamp et at 2006, supra, and the references cited therein).
  • Antigen induced arthritis model can be used to test ADAM-TS5 Nanobodies. (ADAM-TS5-/- mice are more resistant to arthritis in this model).
  • An ELIS A-based kit can be purchased from Invitek to test the activity of amino acid sequences or polypeptides of the invention on ADAM-TS5.
  • cleavage of CD44 into the medium of cell can be used to detect the activity of ADAMlO in U251 glioblastoma (S. Atkinson and G. Murphy, publication submitted).
  • the release of HB-EGF conjugated Alkaline phosphatase from transfected MCF7 cells can be used a readout for ADAM 17 activity (S. Atkinson and G. Murphy, submitted).
  • amino acid sequences and polypeptides that are directed against a metalloproteinase from the ADAM family from a first species of warmblooded animal may or may not show cross-reactivity with a metalloproteinase from the ADAM family from one or more other species of warm-blooded animal.
  • amino acid sequences and polypeptides directed against human a metalloproteinase from the ADAM family may or may not show cross reactivity with a metalloproteinase from the ADAM family from one or more other species of primates (such as, without limitation, monkeys from the genus Macaca (such as, and in particular, cynomolgus monkeys ⁇ Macaca fascicularis) and/or rhesus monkeys ⁇ Macaca mulatto)) and baboon (Papio ursinus)) and/or with a metalloproteinase from the ADAM family from one or more species of animals that are often used in animal models for diseases (for example mouse, rat, rabbit, pig or dog), and in particular in animal models for diseases and disorders associated with a metalloproteinase from the ADAM family (such as the species and animal models mentioned herein).
  • primates such as, without limitation, monkeys from the genus Macaca (such as, and in particular,
  • cross-reactivity when present, may have advantages from a drug development point of view, since it allows the amino acid sequences and polypeptides against human a metalloproteinase from the ADAM family to be tested in such disease models. More generally, amino acid sequences and polypeptides of the invention that are cross-reactive with a metalloproteinase from the ADAM family from multiple species of mammal will usually be advantageous for use in veterinary applications, since it will allow the same amino acid sequence or polypeptide to be used across multiple species.
  • amino acid sequences and polypeptides directed against a metalloproteinase from the ADAM family from one species of animal can be used in the treatment of another species of animal, as long as the use of the amino acid sequences and/or polypeptides provide the desired effects in the species to be treated.
  • the present invention is in its broadest sense also not particularly limited to or defined by a specific antigenic determinant, epitope, part, domain, subunit or confirmation (where applicable) of a metalloproteinase from the ADAM family against which the amino acid sequences and polypeptides of the invention are directed.
  • the amino acid sequences and polypeptides may or may not be directed against an "interaction site" (as defined herein).
  • amino acid sequences of the invention may be directed against one or more of the following epitopes, domains, active/catalytic sites or binding sites: against the catalytic site: such amino acid sequences may for example reduce, inhibit or inactivate the protease activity; against the catalytic domain and/or the PRO-domain-catalytic site (such that it is disrupted): such interaction may for example activate or increase the protease activity; against the PRO-domain and /or against the PRO -catalytic site (such that it is disrupted): such interaction may for example activate or increase the protease activity; - against the Disintegrin/cys-rich domain: such interaction may for example modulate the targeting of the ADAM or its binding to substrate, or its selectivity for the substrate; against the site on the ADAM proteinase for interaction with TIMP, thus modulating (and in particular inhibiting) the regulation of ADAM by TIMPs (TIMP1,2,3 or 4).
  • some preferred amino acid sequences and polypeptides of the invention are preferably such that they are capable of inhibiting the activity of the ADAM proteinase, as measured by a suitable assay (such as a commercially available protease assay) under conditions usually applied for such an assay (as will be clear to the skilled person), by at least 1%, preferably at least 5%, more preferably at least 10%, such as at least 25% or even more than 50% and up to 75% or more, such as 90% or more, compared to the activity of the ADAM proteinase without the presence of the amino acid sequence or polypeptide of the invention.
  • a suitable assay such as a commercially available protease assay
  • some preferred amino acid sequences and polypeptides of the invention are preferably such that they are capable of activating (the protease activity of) the ADAM proteinase, as measured by a suitable assay (such as a commercially available protease assay) under conditions usually applied for such an assay (as will be clear to the skilled person), by at least 1%, preferably at least 5%, more preferably at least 10%, such as at least 25% or even more than 50% and up to 75% or more, such as 90% or more, compared to the activity of the ADAM proteinase without the presence of the amino acid sequence or polypeptide of the invention.
  • a suitable assay such as a commercially available protease assay
  • Some other preferred amino acid sequences and polypeptides may be such that they are capable of competing for binding to the ADAM proteinase with the usual substrate of the ADAM proteinase, and/or with a substrate for an ADAM proteinase that is usually used as a model substrate in an assay for determining the activity of said proteinase.
  • Some other preferred amino acid sequences and polypeptides may be such that they are capable of modulating (as defined herein) the regulation of the ADAM proteinase by TIMP.
  • therapies involving the use of TIMP or targeting may lead to an undesirable inhibition of metalloproteases, which may cause side effects.
  • one aspect of the invention relates to the use of an amino acid sequence, Nanobody or polypeptide of the invention, or of a composition comprising the same (methods or compositions for) preventing or reducing the side effects of TIMP-dependent therapies or therapies directed against or mediated by TIMP; and to uses of the amino acid sequences, Nanobodies and polypeptides of the invention in the preparation of pharmaceutical compositions for reducing the side effects of TIMP-dependent therapies or therapies directed against or mediated by TIMP; or with such reduced side effects.
  • the invention also relates to the use of an amino acid sequence, Nanobody or polypeptide of the invention, or of a composition comprising the same, in (methods or compositions for) modulating (as defined herein) an ADAM metalloproteinase, either in vitro (e.g. in an in vitro or cellular assay) or in vivo (e.g. in an a single cell or in a multicellular organism, and in particular in a mammal, and more in particular in a human being, such as in a human being that is at risk of or suffers from ADAM -related diseases and disorders).
  • the invention also relates to methods for modulating (as defined herein) an ADAM metalloproteinase, either in vitro (e.g.
  • method comprises at least the step of contacting an ADAM metalloproteinase with at least one amino acid sequence, Nanobody or polypeptide of the invention, or with a composition comprising the same, in a manner and in an amount suitable to modulate an ADAM metalloproteinase, with at least one amino acid sequence, Nanobody or polypeptide of the invention.
  • the invention also relates to the use of an one amino acid sequence, Nanobody or polypeptide of the invention in the preparation of a composition (such as, without limitation, a pharmaceutical composition or preparation as further described herein) for modulating (as defined herein) an ADAM metalloproteinase, either in vitro (e.g. in an in vitro or cellular assay) or in vivo (e.g. in an a single cell or multicellular organism, and in particular in a mammal, and more in particular in a human being, such as in a human being that is at risk of or suffers from a ADAM -related diseases and disorders).
  • a composition such as, without limitation, a pharmaceutical composition or preparation as further described herein
  • modulating as defined herein
  • an ADAM metalloproteinase either in vitro (e.g. in an in vitro or cellular assay) or in vivo (e.g. in an a single cell or multicellular organism, and in particular in a mammal, and more
  • amino acid sequences, Nanobodies and polypeptides of the invention may also be in the prevention and treatment of sepsis.
  • further aspects of the invention relate to the use of an amino acid sequence, Nanobody or polypeptide of the invention, or of a composition comprising the same, in (methods or compositions for) the treatment (as defined herein) of sepsis; to pharmaceutical compositions and preparations (as described herein) for the treatment of sepsis; and to the use of the amino acid sequences, Nanobodies and polypeptides of the invention in methods for preparing pharmaceutical compositions and preparations (as described herein) for the treatment of sepsis.
  • the amino acid sequences, Nanobodies and polypeptides of the invention may also be used in the prevention and treatment of the side effects of TIMP-related therapies.
  • further aspects of the invention relate to the use of an amino acid sequence, Nanobody or polypeptide of the invention, or of a composition comprising the same, in (methods or compositions for) the prevention and/or treatment (as defined herein) of the side effects of TIMP-related therapies; to pharmaceutical compositions and preparations (as described herein) for the prevention and/or treatment (as defined herein) of the side effects of TIMP- related therapies; and to the use of the amino acid sequences, Nanobodies and polypeptides of the invention in methods for preparing pharmaceutical compositions and preparations (as described herein) for the prevention and/or treatment (as defined herein) of the side effects of TIMP-related therapies.
  • the amino acid sequences, Nanobodies and polypeptides of the invention may also be used to inhibit or reduce the release of one or more growth factors, for example in the treatment of cancer.
  • further aspects of the invention relate to the use of an amino acid sequence, Nanobody or polypeptide of the invention, or of a composition comprising the same, in (methods or compositions for) inhibiting or reducing the release of one or more growth factors, for example in the treatment of cancer; to pharmaceutical compositions and preparations (as described herein) for inhibiting or reducing the release of one or more growth factors, for example in the treatment of cancer; and to the use of the amino acid sequences, Nanobodies and polypeptides of the invention in methods for preparing pharmaceutical compositions and preparations (as described herein) for inhibiting or reducing the release of one or more growth factors, for example in the treatment of cancer.
  • the amino acid sequences, Nanobodies and polypeptides of the invention may also be used to inhibit or reduce the release of one or more cytokines, for example in the prevention or treatment of inflammation.
  • further aspects of the invention relate to the use of an amino acid sequence, Nanobody or polypeptide of the invention, or of a composition comprising the same, in (methods or compositions for) inhibiting or reducing the release of one or more cytokines, for example in the prevention or treatment of inflammation; to pharmaceutical compositions and preparations (as described herein) for inhibiting or reducing the release of one or more cytokines, for example in the prevention or treatment of inflammation; and to the use of the amino acid sequences, Nanobodies and polypeptides of the invention in methods for preparing pharmaceutical compositions and preparations (as described herein) for inhibiting or reducing the release of one or more cytokines, for example in the prevention or treatment of inflammation.
  • Nanobodies and polypeptides of the invention may also be used as a marker for cells that express ADAM proteinases (such as ADAM 15) on their surface, for example for detecting tumour cells or atherosclerosis, for example in vitro or by means of in vivo imaging.
  • ADAM proteinases such as ADAM 15
  • a polypeptide of the invention may contain two or more amino acid sequences of the invention that are directed against an ADAM proteinase. Generally, such polypeptides will bind to the ADAM proteinase with increased avidity compared to a single amino acid sequence of the invention.
  • Such a polypeptide may for example comprise two amino acid sequences of the invention that are directed against the same antigenic determinant, epitope, part, domain, subunit or confirmation (where applicable) of ADAM proteinase (which may or may not be an interaction site); or comprise at least one "first" amino acid sequence of the invention that is directed against a first same antigenic determinant, epitope, part, domain, subunit or confirmation (where applicable) of ADAM proteinase (which may or may not be an interaction site); and at least one "second” amino acid sequence of the invention that is directed against a second antigenic determinant, epitope, part, domain, subunit or confirmation (where applicable) different from the first (and which again may or may not be an interaction site).
  • at least one amino acid sequence of the invention is directed against an interaction site (as defined herein), although the invention in its broadest sense is not limited thereto.
  • the amino acid sequences and polypeptides may be such that they compete with the cognate binding partner (e.g. the ligand, receptor or other binding partner, as applicable) for binding to the target, and/or such that they (fully or partially) neutralize binding of the binding partner to the target.
  • the cognate binding partner e.g. the ligand, receptor or other binding partner, as applicable
  • an amino acid sequence of the invention can bind to two or more antigenic determinants, epitopes, parts, domains, subunits or confirmations of a metalloproteinase from the ADAM family.
  • the antigenic determinants, epitopes, parts, domains or subunits of a metalloproteinase from the ADAM family to which the amino acid sequences and/or polypeptides of the invention bind may be essentially the same (for example, if a metalloproteinase from the ADAM family contains repeated structural motifs or occurs in a multimeric form) or may be different (and in the latter case, the amino acid sequences and polypeptides of the invention may bind to such different antigenic determinants, epitopes, parts, domains, subunits of a metalloproteinase from the ADAM family with an affinity and/or specificity which may be the same or different).
  • amino acid sequences and polypeptides of the invention may bind to either one of these confirmation, or may bind to both these confirmations (i.e. with an affinity and/or specificity which may be the same or different).
  • amino acid sequences and polypeptides of the invention may bind to a conformation of a metalloproteinase from the ADAM family in which it is bound to a pertinent ligand, may bind to a conformation of a metalloproteinase from the ADAM family in which it not bound to a pertinent ligand, or may bind to both such conformations (again with an affinity and/or specificity which may be the same or different).
  • amino acid sequences and polypeptides of the invention will generally bind to all naturally occurring or synthetic analogs, variants, mutants, alleles, parts and fragments of a metalloproteinase from the ADAM family; or at least to those analogs, variants, mutants, alleles, parts and fragments of a metalloproteinase from the ADAM family that contain one or more antigenic determinants or epitopes that are essentially the same as the antigenic determinant(s) or epitope(s) to which the amino acid sequences and polypeptides of the invention bind in a metalloproteinase from the ADAM family (e.g. in wild-type a metalloproteinase from the ADAM family).
  • the amino acid sequences and polypeptides of the invention may bind to such analogs, variants, mutants, alleles, parts and fragments with an affinity and/or specificity that are the same as, or that are different from (i.e. higher than or lower than), the affinity and specificity with which the amino acid sequences of the invention bind to (wild-type) a metalloproteinase from the ADAM family. It is also included within the scope of the invention that the amino acid sequences and polypeptides of the invention bind to some analogs, variants, mutants, alleles, parts and fragments of a metalloproteinase from the ADAM family, but not to others.
  • the amino acid sequences and polypeptides of the invention only bind to a metalloproteinase from the ADAM family in monomeric form, only bind to a metalloproteinase from the ADAM family in multimeric form, or bind to both the monomeric and the multimeric form.
  • the amino acid sequences and polypeptides of the invention may bind to the monomeric form with an affinity and/or specificity that are the same as, or that are different from (i.e. higher than or lower than), the affinity and specificity with which the amino acid sequences of the invention bind to the multimeric form.
  • amino acid sequences and polypeptides of the invention bind to a metalloproteinase from the ADAM family in its non-associated state, bind to a metalloproteinase from the ADAM family in its associated state, or bind to both.
  • the amino acid sequences and polypeptides of the invention may bind to such multimers or associated protein complexes with an affinity and/or specificity that may be the same as or different from (i.e.
  • proteins or polypeptides that contain two or more amino acid sequences directed against a metalloproteinase from the ADAM family may bind with higher avidity to a metalloproteinase from the ADAM family than the corresponding monomeric amino acid sequence(s).
  • proteins or polypeptides that contain two or more amino acid sequences directed against different epitopes of a metalloproteinase from the ADAM family may (and usually will) bind with higher avidity than each of the different monomers, and proteins or polypeptides that contain two or more amino acid sequences directed against a metalloproteinase from the ADAM family may (and usually will) bind also with higher avidity to a multimer of a metalloproteinase from the ADAM family.
  • amino acid sequences and polypeptides of the invention will at least bind to those forms of a metalloproteinase from the ADAM family (including monomeric, multimeric and associated forms) that are the most relevant from a biological and/or therapeutic point of view, as will be clear to the skilled person.
  • Such parts, fragments, analogs, mutants, variants, alleles and/or derivatives will usually contain (at least part of) a functional antigen-binding site for binding against a metalloproteinase from the ADAM family; and more preferably will be capable of specific binding to a metalloproteinase from the ADAM family, and even more preferably capable of binding to a metalloproteinase from the ADAM family with an affinity (suitably measured and/or expressed as a K ⁇ -value (actual or apparent), a K A -value (actual or apparent), a k on -rate and/or a k off -rate, or alternatively as an IC 5O value, as further described herein) that is as defined herein.
  • fragments or polypeptides of the invention may also be provided by suitably combining (i.e. by linking or genetic fusion) one or more (smaller) parts or fragments as described herein.
  • analogs, mutants, variants, alleles, derivatives have an increased half- life in serum (as further described herein) compared to the amino acid sequence from which they have been derived.
  • an amino acid sequence of the invention may be linked (chemically or otherwise) to one or more groups or moieties that extend the half-life (such as PEG), so as to provide a derivative of an amino acid sequence of the invention with increased half-life.
  • the amino acid sequence of the invention may be an amino acid sequence that comprises an immunoglobulin fold or may be an amino acid sequence that, under suitable conditions (such as physiological conditions) is capable of forming an immunoglobulin fold (i.e. by folding).
  • suitable conditions such as physiological conditions
  • such an amino acid sequence when properly folded so as to form an immunoglobulin fold, is capable of specific binding (as defined herein) to a metalloproteinase from the ADAM family; and more preferably capable of binding to a metalloproteinase from the ADAM family with an affinity (suitably measured and/or expressed as a K ⁇ -value (actual or apparent), a KA-value (actual or apparent), a kon- rate and/or a k off -rate, or alternatively as an IC 5O value, as further described herein) that is as defined herein.
  • parts, fragments, analogs, mutants, variants, alleles and/or derivatives of such amino acid sequences are preferably such that they comprise an immunoglobulin fold or are capable for forming, under suitable conditions, an immunoglobulin fold.
  • amino acid sequences of the invention may be amino acid sequences that essentially consist of 4 framework regions (FRl to FR4 respectively) and 3 complementarity determining regions (CDRl to CDR3 respectively); or any suitable fragment of such an amino acid sequence (which will then usually contain at least some of the amino acid residues that form at least one of the CDR's, as further described herein).
  • the amino acid sequences of the invention may in particular be an immunoglobulin sequence or a suitable fragment thereof, and more in particular be an immunoglobulin variable domain sequence or a suitable fragment thereof, such as light chain variable domain sequence (e.g. a V L -sequence) or a suitable fragment thereof; or a heavy chain variable domain sequence (e.g. a V H -sequence) or a suitable fragment thereof.
  • an immunoglobulin variable domain sequence or a suitable fragment thereof such as light chain variable domain sequence (e.g. a V L -sequence) or a suitable fragment thereof; or a heavy chain variable domain sequence (e.g. a V H -sequence) or a suitable fragment thereof.
  • the amino acid sequence of the invention when it is a heavy chain variable domain sequence, it may be a heavy chain variable domain sequence that is derived from a conventional four-chain antibody (such as, without limitation, a V H sequence that is derived from a human antibody) or be a so-called VHH-sequence (as defined herein) that is derived from a so-called “heavy chain antibody” (as defined herein).
  • a conventional four-chain antibody such as, without limitation, a V H sequence that is derived from a human antibody
  • VHH-sequence as defined herein
  • the invention is not limited as to the origin of the amino acid sequence of the invention (or of the nucleotide sequence of the invention used to express it), nor as to the way that the amino acid sequence or nucleotide sequence of the invention is (or has been) generated or obtained.
  • the amino acid sequences of the invention may be naturally occurring amino acid sequences (from any suitable species) or synthetic or semi-synthetic amino acid sequences.
  • the amino acid sequence is a naturally occurring immunoglobulin sequence (from any suitable species) or a synthetic or semi-synthetic immunoglobulin sequence, including but not limited to "humanized” (as defined herein) immunoglobulin sequences (such as partially or fully humanized mouse or rabbit immunoglobulin sequences, and in particular partially or fully humanized V HH sequences or Nanobodies), "camelized” (as defined herein) immunoglobulin sequences, as well as immunoglobulin sequences that have been obtained by techniques such as affinity maturation (for example, starting from synthetic, random or naturally occurring immunoglobulin sequences), CDR grafting, veneering, combining fragments derived from different immunoglobulin sequences, PCR assembly using overlapping primers, and similar techniques for engineering immunoglobulin sequences well known to the skilled person; or any suitable combination of any of the foregoing.
  • “humanized” as defined herein
  • immunoglobulin sequences such as partially or fully humanized mouse or rabbit immunoglobulin sequences, and in particular
  • nucleotide sequences of the invention may be naturally occurring nucleotide sequences or synthetic or semi-synthetic sequences, and may for example be sequences that are isolated by PCR from a suitable naturally occurring template (e.g.
  • DNA or RNA isolated from a cell DNA or RNA isolated from a cell
  • nucleotide sequences that have been isolated from a library and in particular, an expression library
  • nucleotide sequences that have been prepared by introducing mutations into a naturally occurring nucleotide sequence using any suitable technique known per se, such as mismatch PCR
  • nucleotide sequence that have been prepared by PCR using overlapping primers or nucleotide sequences that have been prepared using techniques for DNA synthesis known per se.
  • the amino acid sequence of the invention may in particular be a domain antibody (or an amino acid sequence that is suitable for use as a domain antibody), a single domain antibody (or an amino acid sequence that is suitable for use as a single domain antibody), a "dAb” (or an amino acid sequence that is suitable for use as a dAb) or aNanobodyTM (as defined herein, and including but not limited to a VHH sequence); other single variable domains, or any suitable fragment of any one thereof.
  • a domain antibody or an amino acid sequence that is suitable for use as a domain antibody
  • a single domain antibody or an amino acid sequence that is suitable for use as a single domain antibody
  • a “dAb” or an amino acid sequence that is suitable for use as a dAb
  • aNanobodyTM as defined herein, and including but not limited to a VHH sequence
  • the amino acid sequence of the invention may be a NanobodyTM (as defined herein) or a suitable fragment thereof.
  • Nanobodies of the invention are registered trademarks ofAbfynx N VJ Such Nanobodies directed against a metalloproteinase from the ADAM family will also be referred to herein as "Nanobodies of the invention'.
  • Nanobodies of the invention' For a general description of Nanobodies, reference is made to the further description below, as well as to the prior art cited herein. In this respect, it should however be noted that this description and the prior art mainly described Nanobodies of the so-called "V H 3 class" (i.e.
  • Nanobodies with a high degree of sequence homology to human germline sequences of the VH3 class such as DP-47, DP-51 or DP-29
  • the invention in its broadest sense generally covers any type of Nanobody directed against a metalloproteinase from the ADAM family, and for example also covers the Nanobodies belonging to the so-called "V H 4 class” (i.e. Nanobodies with a high degree of sequence homology to human germline sequences of the V H 4 class such as DP-78), as for example described in the US provisional application 60/792,279 by Ablynx N.V. entitled “DP-78-like Nanobodies” filed on April 14, 2006 (see also PCT/EP2007/003259).
  • Nanobodies in particular V HH sequences and partially humanized Nanobodies
  • V HH sequences and partially humanized Nanobodies can in particular be characterized by the presence of one or more "Hallmark residues" (as described herein) in one or more of the framework sequences (again as further described herein).
  • Nanobody can be defined as an amino acid sequence with the (general) structure
  • FRl to FR4 refer to framework regions 1 to 4, respectively, and in which CDRl to CDR3 refer to the complementarity determining regions 1 to 3, respectively, and in which one or more of the Hallmark residues are as further defined herein.
  • Nanobody can be an amino acid sequence with the (general) structure
  • FRl to FR4 refer to framework regions 1 to 4, respectively, and in which CDRl to CDR3 refer to the complementarity determining regions 1 to 3, respectively, and in which the framework sequences are as further defined herein.
  • Nanobody can be an amino acid sequence with the (general) structure
  • FRl to FR4 refer to framework regions 1 to 4, respectively, and in which CDRl to CDR3 refer to the complementarity determining regions 1 to 3, respectively, and in which: a) preferably one or more of the amino acid residues at positions 11, 37, 44, 45, 47, 83,
  • the invention also relates to such Nanobodies that can bind to (as defined herein) and/or are directed against a metalloproteinase from the ADAM family, to suitable fragments thereof, as well as to polypeptides that comprise or essentially consist of one or more of such Nanobodies and/or suitable fragments.
  • SEQ ID NO's 868 to 973 and 1044 to 1053 give the amino acid sequences of a number of V HH sequences that have been raised against a metalloproteinase from the ADAM family, i.e.:
  • SEQ ID NO's 868 to 886 Nanobodies against ADAM8 - SEQ ID NO's 887 to 907 and 1044 to 1047: Nanobodies against ADAM9 SEQ ID NO's 908 to 931 : Nanobodies against ADAMlO SEQ ID NO's 932 to 948 and 1048 to 1052: Nanobodies against ADAM17 SEQ ID NO's 949 to 974 and 1053: Nanobodies against ADAMTS5
  • the invention in some specific aspects provides: - amino acid sequences that are directed against (as defined herein) ADAM8 and that have at least 80%, preferably at least 85%, such as 90% or 95% or more sequence identity with at least one of the amino acid sequences of SEQ ID NO's: 868 to 886.
  • These amino acid sequences may further be such that they neutralize binding of the cognate ligand to ADAM8; and/or compete with the cognate ligand for binding to ADAM8; and/or are directed against an interaction site (as defined herein) on ADAM8
  • amino acid sequences that cross-block (as defined herein) the binding of at least one of the amino acid sequences of SEQ ID NO's: 868 to 886 to ADAM8 and/or that compete with at least one of the amino acid sequences of SEQ ID NO's: 868 to 886 for binding to ADAM8.
  • these amino acid sequences may further be such that they neutralize binding of the cognate ligand to ADAM8; and/or compete with the cognate ligand for binding to ADAM8; and/or are directed against an interaction site (as defined herein) on ADAM8 (such as the ligand binding site);
  • amino acid sequences that are directed against (as defined herein) ADAM9 and that have at least 80%, preferably at least 85%, such as 90% or 95% or more sequence identity with at least one of the amino acid sequences of SEQ ID NO's: 887 to 907 and 1044 to 1047.
  • amino acid sequences may further be such that they neutralize binding of the cognate ligand to ADAM9; and/or compete with the cognate ligand for binding to ADAM9; and/or are directed against an interaction site (as defined herein) on ADAM9 (such as the ligand binding site);
  • amino acid sequences that cross-block (as defined herein) the binding of at least one of the amino acid sequences of SEQ ID NO's: 887 to 907 and 1044 to 1047to ADAM9 and/or that compete with at least one of the amino acid sequences of SEQ ID NO's: 887 to 907 and 1044 to 1047 for binding to ADAM9.
  • these amino acid sequences may further be such that they neutralize binding of the cognate ligand to ADAM9; and/or compete with the cognate ligand for binding to ADAM9; and/or are directed against an interaction site (as defined herein) on ADAM9 (such as the ligand binding site);
  • amino acid sequences that are directed against (as defined herein) ADAMlO and that have at least 80%, preferably at least 85%, such as 90% or 95% or more sequence identity with at least one of the amino acid sequences of SEQ ID NO's: 908 to 931. These amino acid sequences may further be such that they neutralize binding of the cognate ligand to ADAMlO; and/or compete with the cognate ligand for binding to
  • ADAMlO and/or are directed against an interaction site (as defined herein) on ADAMlO (such as the ligand binding site);
  • these amino acid sequences may further be such that they neutralize binding of the cognate ligand to ADAMlO; and/or compete with the cognate ligand for binding to ADAMlO; and/or are directed against an interaction site (as defined herein) on ADAMlO (such as the ligand binding site); - amino acid sequences that are directed against (as defined herein) ADAM 17 and that have at least 80%, preferably at least 85%, such as 90% or 95% or more sequence identity with at least one of the amino acid sequences of SEQ ID NO's: 932 to 948 and 1048 to 1052.
  • amino acid sequences may further be such that they neutralize binding of the cognate ligand to ADAM17; and/or compete with the cognate ligand for binding to ADAM 17; and/or are directed against an interaction site (as defined herein) on ADAM 17 (such as the ligand binding site);
  • amino acid sequences that cross-block (as defined herein) the binding of at least one of the amino acid sequences of SEQ ID NO's: 932 to 948 and 1048 to 1052to ADAM17 and/or that compete with at least one of the amino acid sequences of SEQ ID NO's: 932 to 948 and 1048 to 1052 for binding to ADAMl 7.
  • these amino acid sequences may further be such that they neutralize binding of the cognate ligand to ADAM17; and/or compete with the cognate ligand for binding to ADAM 17; and/or are directed against an interaction site (as defined herein) on ADAM 17 (such as the ligand binding site);
  • amino acid sequences that are directed against (as defined herein) ADAMT S 5 and that have at least 80%, preferably at least 85%, such as 90% or 95% or more sequence identity with at least one of the amino acid sequences of SEQ ID NO's: 949 to 974 and 1053.
  • amino acid sequences may further be such that they neutralize binding of the cognate ligand to ADAMTS5; and/or compete with the cognate ligand for binding to ADAMTS5; and/or are directed against an interaction site (as defined herein) on ADAMTS5 (such as the ligand binding site);
  • amino acid sequences may further be such that they neutralize binding of the cognate ligand to ADAMTS5; and/or compete with the cognate ligand for binding to ADAMTS5; and/or are directed against an interaction site (as defined herein) on ADAMTS5 (such as the ligand binding site); which amino acid sequences may be as further described herein (and may for example be Nanobodies); as well as polypeptides of the invention that comprise one or more of such amino acid sequences (which may be as further described herein, and may for example be bispecific and/or biparatopic polypeptides as described herein), and nucleic acid sequences that encode such amino acid sequences and polypeptides. Such amino acid sequences and polypeptides do not include any naturally occurring ligands.
  • the invention provides:
  • ADAM8 compared to ADAM9, ADAMlO, ADAM 17 and/or ADAMTS5.
  • ADAM8 compared to ADAM9, ADAMlO, ADAM 17 and/or ADAMTS5.
  • ADAMTS5 ADAMTS5.
  • These preferably have have at least 80%, preferably at least 85%, such as 90% or 95% or more sequence identity with at least one of the amino acid sequences of SEQ ID NO's: 868 to 886;
  • ADAM9 compared to ADAM8, ADAMlO, ADAM17 and/or ADAMTS5.
  • ADAM9 compared to ADAM8, ADAMlO, ADAM17 and/or ADAMTS5.
  • These preferably have at least 80%, preferably at least 85%, such as 90% or 95% or more sequence identity with at least one of the amino acid sequences of SEQ ID NO's: 887 to 907 and 1044 to 1047.
  • ADAMlO compared to ADAM8, ADAM9, ADAM17 and/or ADAMTS5.
  • ADAMlO compared to ADAM8, ADAM9, ADAM17 and/or ADAMTS5.
  • These preferably have at least 80%, preferably at least 85%, such as 90% or 95% or more sequence identity with at least one of the amino acid sequences of SEQ ID NO's: 908 to 931;
  • ADAM17 compared to ADAM8, ADAM9, ADAMlO and/or ADAMTS5.
  • ADAM17 compared to ADAM8, ADAM9, ADAMlO and/or ADAMTS5.
  • These preferably have at least 80%, preferably at least 85%, such as 90% or 95% or more sequence identity with at least one of the amino acid sequences of SEQ ID NO's: 932 to 948 and 1048 to 1052;
  • amino acid sequences of the invention that are specific for (as defined herein) ADAMTS5 compared to ADAM8, ADAM9, ADAMlO and/or ADAM17. These preferably have at least 80%, preferably at least 85%, such as 90% or 95% or more sequence identity with at least one of the amino acid sequences of SEQ ID NO's: 949 to 974 and 1053; which amino acid sequences of the invention may be as further described herein (and may for example be Nanobodies); as well as polypeptides of the invention that comprise one or more of such amino acid sequences (which may be as further described herein, and may for example be bispecific and/or biparatopic polypeptides as described herein), and nucleic acid sequences that encode such amino acid sequences and polypeptides.
  • Nanobodies of the invention are Nanobodies which can bind (as further defined herein) to and/or are directed against to a metalloproteinase from the ADAM family and which: a) have at least 80% amino acid identity with at least one of the amino acid sequences of
  • these residues are preferably disregarded); and in which: b) preferably one or more of the amino acid residues at positions 11, 37, 44, 45, 47, 83, 84, 103, 104 and 108 according to the Kabat numbering are chosen from the Hallmark residues mentioned in Table A-3 below.
  • the CDR sequences are generally as further defined herein.
  • Nanobodies may be derived in any suitable manner and from any suitable source, and may for example be naturally occurring V HH sequences (i.e. from a suitable species of Camelid) or synthetic or semi-synthetic amino acid sequences, including but not limited to "humanized” (as defined herein) Nanobodies, “camelized” (as defined herein) immunoglobulin sequences (and in particular camelized heavy chain variable domain sequences), as well as Nanobodies that have been obtained by techniques such as affinity maturation (for example, starting from synthetic, random or naturally occurring immunoglobulin sequences), CDR grafting, veneering, combining fragments derived from different immunoglobulin sequences, PCR assembly using overlapping primers, and similar techniques for engineering immunoglobulin sequences well known to the skilled person; or any suitable combination of any of the foregoing as further described herein.
  • affinity maturation for example, starting from synthetic, random or naturally occurring immunoglobulin sequences
  • CDR grafting for example, starting from synthetic, random or naturally occurring
  • Nanobody when a Nanobody comprises a V HH sequence, said Nanobody may be suitably humanized, as further described herein, so as to provide one or more further (partially or fully) humanized Nanobodies of the invention.
  • a Nanobody when a Nanobody comprises a synthetic or semi- synthetic sequence (such as a partially humanized sequence), said Nanobody may optionally be further suitably humanized, again as described herein, again so as to provide one or more further (partially or fully) humanized Nanobodies of the invention.
  • humanized Nanobodies may be amino acid sequences that are as generally defined for Nanobodies in the previous paragraphs, but in which at least one amino acid residue is present (and in particular, in at least one of the framework residues) that is and/or that corresponds to a humanizing substitution (as defined herein).
  • a humanizing substitution as defined herein.
  • Some particularly preferred humanized Nanobodies of the invention are humanized variants of the Nanobodies of SEQ ID NO's: 868 to 973 and/or 1044 to 1053.
  • Nanobodies of the invention are Nanobodies which can bind (as further defined herein) to a metalloproteinase from the ADAM family and which: a) are a humanized variant of one of the amino acid sequences of SEQ ID NO's: 868 to 973 and/or 1044 to 1053; and/or b) have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 868 to 973 and/or 1044 to 1053, in which for the purposes of determining the degree of amino acid identity, the amino acid residues that form the
  • CDR sequences are disregarded; and in which: c) preferably one or more of the amino acid residues at positions 11, 37, 44, 45, 47, 83, 84, 103, 104 and 108 according to the Kabat numbering are chosen from the Hallmark residues mentioned in Table A-3 below.
  • the invention provides a number of stretches of amino acid residues (i.e. small peptides) that are particularly suited for binding to a metalloproteinase from the ADAM family. These stretches of amino acid residues may be present in, and/or may be corporated into, an amino acid sequence of the invention, in particular in such a way that they form (part of) the antigen binding site of an amino acid sequence of the invention.
  • CDR sequences ' i.e. as CDRl sequences, CDR2 sequences and CDR3 sequences, respectively.
  • the invention in its broadest sense is not limited to a specific structural role or function that these stretches of amino acid residues may have in an amino acid sequence of the invention, as long as these stretches of amino acid residues allow the amino acid sequence of the invention to bind to a metalloproteinase from the ADAM family.
  • the invention in its broadest sense comprises any amino acid sequence that is capable of binding to a metalloproteinase from the ADAM family and that comprises one or more CDR sequences as described herein, and in particular a suitable combination of two or more such CDR sequences, that are suitably linked to each other via one or more further amino acid sequences, such that the entire amino acid sequence forms a binding domain and/or binding unit that is capable of binding to a metalloproteinase from the ADAM family.
  • the amino acid sequence of the invention may be an amino acid sequence that comprises at least one amino acid sequence that is chosen from the group consisting of the CDRl sequences, CDR2 sequences and CDR3 sequences that are described herein (or any suitable combination thereof).
  • an amino acid sequence of the invention may be an amino acid sequence that comprises at least one antigen binding site, wherein said antigen binding site comprises at least one amino acid sequence that is chosen from the group consisting of the CDRl sequences, CDR2 sequences and CDR3 sequences that are described herein (or any suitable combination thereof).
  • the amino acid sequence of the invention may be any amino acid sequence that comprises at least one stretch of amino acid residues, in which said stretch of amino acid residues has an amino acid sequence that corresponds to the sequence of at least one of the CDR sequences described herein.
  • Such an amino acid sequence may or may not comprise an immunoglobulin fold.
  • such an amino acid sequence may be a suitable fragment of an immunoglobulin sequence that comprises at least one such CDR sequence, but that is not large enough to form a (complete) immunoglobulin fold (reference is for example again made to the "Expedite fragments" described in WO 03/050531).
  • such an amino acid sequence may be a suitable "protein scaffold” that comprises least one stretch of amino acid residues that corresponds to such a CDR sequence (i.e. as part of its antigen binding site).
  • Suitable scaffolds for presenting amino acid sequences will be clear to the skilled person, and for example comprise, without limitation, to binding scaffolds based on or derived from immunoglobulins (i.e. other than the immunoglobulin sequences already described herein), protein scaffolds derived from protein A domains (such as AffibodiesTM), tendamistat, fibronectin, lipocalin, CTLA-4, T-cell receptors, designed ankyrin repeats, avimers and PDZ domains (Binz et al., Nat. Biotech 2005, VoI 23:1257), and binding moieties based on DNA or RNA including but not limited to DNA or RNA aptamers (Ulrich et al., Comb Chem High Throughput Screen 2006 9(8):619-32).
  • any amino acid sequence of the invention that comprises one or more of these CDR sequences is preferably such that it can specifically bind (as defined herein) to a metalloproteinase from the ADAM family, and more in particular such that it can bind to a metalloproteinase from the ADAM family with an affinity (suitably measured and/or expressed as a K D -value (actual or apparent), a K A -value (actual or apparent), a k on -rate and/or a k ofr rate, or alternatively as an IC 50 value, as further described herein), that is as defined herein.
  • the amino acid sequences according to this aspect of the invention may be any amino acid sequence that comprises at least one antigen binding site, wherein said antigen binding site comprises at least two amino acid sequences that are chosen from the group consisting of the CDRl sequences described herein, the CDR2 sequences described herein and the CDR3 sequences described herein, such that (i) when the first amino acid sequence is chosen from the CDRl sequences described herein, the second amino acid sequence is chosen from the CDR2 sequences described herein or the CDR3 sequences described herein; (ii) when the first amino acid sequence is chosen from the CDR2 sequences described herein, the second amino acid sequence is chosen from the CDRl sequences described herein or the CDR3 sequences described herein; or (iii) when the first amino acid sequence is chosen from the CDR3 sequences described herein, the second amino acid sequence is chosen from the CDRl sequences described herein or the CDR3 sequences described herein.
  • the amino acid sequences of the invention may be amino acid sequences that comprise at least one antigen binding site, wherein said antigen binding site comprises at least three amino acid sequences that are chosen from the group consisting of the CDRl sequences described herein, the CDR2 sequences described herein and the CDR3 sequences described herein, such that the first amino acid sequence is chosen from the CDRl sequences described herein, the second amino acid sequence is chosen from the CDR2 sequences described herein, and the third amino acid sequence is chosen from the CDR3 sequences described herein.
  • Preferred combinations of CDRl, CDR2 and CDR3 sequences will become clear from the further description herein.
  • such an amino acid sequence is preferably an immunoglobulin sequence (as further described herein), but it may for example also be any other amino acid sequence that comprises a suitable scaffold for presenting said CDR sequences.
  • the invention relates to an amino acid sequence directed against a metalloproteinase from the ADAM family, that comprises one or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 337; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 232 to 337; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 232 to 337; d) the amino acid sequences of SEQ ID NO's: 444 to 549; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 444 to 549; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's
  • an amino acid sequence of the invention contains one or more amino acid sequences according to b) and/or c): i) any amino acid substitution in such an amino acid sequence according to b) and/or c) is preferably, and compared to the corresponding amino acid sequence according to a), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to b) and/or c) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to a); and/or iii) the amino acid sequence according to b) and/or c) may be an amino acid sequence that is derived from an amino acid sequence according to a) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • an amino acid sequence of the invention contains one or more amino acid sequences according to e) and/or f): i) any amino acid substitution in such an amino acid sequence according to e) and/or f) is preferably, and compared to the corresponding amino acid sequence according to d), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to e) and/or f) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to d); and/or iii) the amino acid sequence according to e) and/or f) may be an amino acid sequence that is derived from an amino acid sequence according to d) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • an amino acid sequence of the invention contains one or more amino acid sequences according to h) and/or i): i) any amino acid substitution in such an amino acid sequence according to h) and/or i) is preferably, and compared to the corresponding amino acid sequence according to g), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to h) and/or i) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to g); and/or iii) the amino acid sequence according to h) and/or i) may be an amino acid sequence that is derived from an amino acid sequence according to g) by means of affinity maturation using one or more techniques of affinity maturation known per se. It should be understood that the last preceding paragraphs also generally apply to any amino acid sequences of the invention that comprise one or more amino acid sequences according to b), c), e), f), h
  • the amino acid sequence preferably comprises one or more stretches of amino acid residues chosen from the group consisting of: i) the amino acid sequences of SEQ ID NO's: 232 to 337; ii) the amino acid sequences of SEQ ID NO's: 444 to 549; and iii) the amino acid sequences of SEQ ID NO's: 656 to 761; or any suitable combination thereof.
  • At least one of said stretches of amino acid residues forms part of the antigen binding site for binding against a metalloproteinase from the ADAM family.
  • the invention relates to an amino acid sequence directed against a metalloproteinase from the ADAM family, that comprises two or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 337; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 232 to 337; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 232 to 337; d) the amino acid sequences of SEQ ID NO's: 444 to 549; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 444 to 549; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO'
  • the amino acid sequence preferably comprises two or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 337; b) the amino acid sequences of SEQ ID NO's: 444 to 549; and c) the amino acid sequences of SEQ ID NO's: 656 to 761; such that, (i) when the first stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 232 to 337, the second stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 444 to 549 or of SEQ ID NO's: 656 to 761; (ii) when the first stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 444 to 549, the second stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 232 to 337 or of SEQ ID
  • the at least two stretches of amino acid residues again preferably form part of the antigen binding site for binding against a metalloproteinase from the ADAM family.
  • the invention relates to an amino acid sequence directed against a metalloproteinase from the ADAM family, that comprises three or more stretches of amino acid residues, in which the first stretch of amino acid residues is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 337; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 232 to 337; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 232 to 337; the second stretch of amino acid residues is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 4
  • the first stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 232 to 337; the second stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 444 to 549; and the third stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 656 to 761.
  • the at least three stretches of amino acid residues forms part of the antigen binding site for binding against a metalloproteinase from the ADAM family.
  • the CDR sequences have at least 70% amino acid identity, preferably at least 80% amino acid identity, more preferably at least 90% amino acid identity, such as 95% amino acid identity or more or even essentially 100% amino acid identity with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 868 to 973 and/or 1044 to 1053.
  • This degree of amino acid identity can for example be determined by determining the degree of amino acid identity (in a manner described herein) between said amino acid sequence and one or more of the sequences of SEQ ID NO's: 868 to 973 and/or 1044 to 1053, in which the amino acid residues that form the framework regions are disregarded.
  • amino acid sequences of the invention can be as further described herein.
  • such amino acid sequences are preferably such that they can specifically bind
  • a metalloproteinase from the ADAM family (as defined herein) to a metalloproteinase from the ADAM family; and more in particular bind to a metalloproteinase from the ADAM family with an affinity (suitably measured and/or expressed as a K ⁇ -value (actual or apparent), a KA-value (actual or apparent), a kon- rate and/or a k off -rate, or alternatively as an IC 5O value, as further described herein) that is as defined herein.
  • the amino acid sequence of the invention is preferably such that: CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 337; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 232 to 337; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 232 to 337; and/or
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 444 to 549; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 444 to 549; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 444 to 549; and/or
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 656 to 761; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 656 to 761; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 656 to 761.
  • such an amino acid sequence of the invention may be such that CDRl is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 232 to 337; and/or CDR2 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 444 to 549; and/or CDR3 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 656 to 761.
  • the amino acid sequence of the invention is preferably such that: - CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 337; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 232 to 337; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 232 to 337; and
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 444 to 549; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 444 to 549; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 444 to 549; and
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 656 to 761; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 656 to 761; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 656 to 761; or any suitable fragment of such an amino acid sequence
  • such an amino acid sequence of the invention may be such that CDRl is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 232 to 337; and CDR2 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 444 to 549; and CDR3 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 656 to 761.
  • amino acid sequences are preferably such that they can specifically bind
  • a metalloproteinase from the ADAM family (as defined herein) to a metalloproteinase from the ADAM family; and more in particular bind to a metalloproteinase from the ADAM family with an affinity (suitably measured and/or expressed as a K ⁇ -value (actual or apparent), a KA-value (actual or apparent), a kon- rate and/or a k off -rate, or alternatively as an IC 5O value, as further described herein) that is as defined herein.
  • the invention relates to an amino acid sequence that essentially consists of 4 framework regions (FRl to FR4, respectively) and 3 complementarity determining regions (CDRl to CDR3, respectively), in which the CDR sequences of said amino acid sequence have at least 70% amino acid identity, preferably at least 80% amino acid identity, more preferably at least 90% amino acid identity, such as 95% amino acid identity or more or even essentially 100% amino acid identity with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 868 to 973 and/or 1044 to 1053.
  • This degree of amino acid identity can for example be determined by determining the degree of amino acid identity (in a manner described herein) between said amino acid sequence and one or more of the sequences of SEQ ID NO's: 868 to 973 and/or 1044 to 1053, in which the amino acid residues that form the framework regions are disregarded.
  • Such amino acid sequences of the invention can be as further described herein.
  • the invention relates to an amino acid sequence directed against ADAM8 (such as aNanobody of the invention, as further described herein), that comprises one or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 250; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 232 to 250; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 232 to 250; d) the amino acid sequences of SEQ ID NO's: 444 to 462; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 444 to 462; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ
  • an amino acid sequence of the invention contains one or more amino acid sequences according to b) and/or c): i) any amino acid substitution in such an amino acid sequence according to b) and/or c) is preferably, and compared to the corresponding amino acid sequence according to a), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to b) and/or c) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to a); and/or iii) the amino acid sequence according to b) and/or c) may be an amino acid sequence that is derived from an amino acid sequence according to a) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • an amino acid sequence of the invention contains one or more amino acid sequences according to e) and/or f): i) any amino acid substitution in such an amino acid sequence according to e) and/or f) is preferably, and compared to the corresponding amino acid sequence according to d), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to e) and/or f) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to d); and/or iii) the amino acid sequence according to e) and/or f) may be an amino acid sequence that is derived from an amino acid sequence according to d) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • an amino acid sequence of the invention contains one or more amino acid sequences according to h) and/or i): i) any amino acid substitution in such an amino acid sequence according to h) and/or i) is preferably, and compared to the corresponding amino acid sequence according to g), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to h) and/or i) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to g); and/or iii) the amino acid sequence according to h) and/or i) may be an amino acid sequence that is derived from an amino acid sequence according to g) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • amino acid sequences of the invention that comprise one or more amino acid sequences according to b), c), e), f), h) or i), respectively.
  • the amino acid sequence preferably comprises one or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 250; b) the amino acid sequences of SEQ ID NO's: 444 to 462; and c) the amino acid sequences of SEQ ID NO's: 656 to 674; or any suitable combination thereof.
  • At least one of said stretches of amino acid residues forms part of the antigen binding site for binding against ADAM8.
  • the invention relates to an amino acid sequence directed against ADAM8, that comprises two or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 250; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 232 to 250; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 232 to 250; d) the amino acid sequences of SEQ ID NO's: 444 to 462; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 444 to 462; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 444 to 462; g) the amino acid sequence directed against ADAM8, that comprises two or more stretches
  • the amino acid sequence preferably comprises two or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 250; b) the amino acid sequences of SEQ ID NO's: 444 to 462; and c) the amino acid sequences of SEQ ID NO's: 656 to 674; such that, (i) when the first stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 232 to 250, the second stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 444 to 462 or of SEQ ID NO's: 656 to 674; (ii) when the first stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 444 to 462, the second stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 232 to 250 or of SEQ ID NO's:
  • the at least two stretches of amino acid residues again preferably form part of the antigen binding site for binding against ADAM8.
  • the invention relates to an amino acid sequence directed against ADAM8, that comprises three or more stretches of amino acid residues, in which the first stretch of amino acid residues is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 250; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 232 to 250; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 232 to 250; the second stretch of amino acid residues is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 444 to 462; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 444 to 462; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least
  • the first stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 232 to 250; the second stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 444 to 462; and the third stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 656 to 674.
  • the at least three stretches of amino acid residues forms part of the antigen binding site for binding against ADAM8. Preferred combinations of such stretches of amino acid sequences will become clear from the further disclosure herein.
  • the CDR sequences have at least 70% amino acid identity, preferably at least 80% amino acid identity, more preferably at least 90% amino acid identity, such as 95% amino acid identity or more or even essentially 100% amino acid identity with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 868 to 886.
  • This degree of amino acid identity can for example be determined by determining the degree of amino acid identity (in a manner described herein) between said amino acid sequence and one or more of the sequences of SEQ ID NO's: 868 to 886, in which the amino acid residues that form the framework regions are disregarded.
  • amino acid sequences of the invention can be as further described herein.
  • amino acid sequences are preferably such that they can specifically bind (as defined herein) to ADAM8; and more in particular bind to ADAM8 with an affinity (suitably measured and/or expressed as a K ⁇ -value (actual or apparent), a KA-value (actual or apparent), a kon-rate and/or a k o ff-rate, or alternatively as an IC50 value, as further described herein) that is as defined herein.
  • the amino acid sequence of the invention is preferably such that: - CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 250; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 232 to 250; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 232 to 250; and/or
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 444 to 462; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 444 to 462; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 444 to 462; and/or CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 656 to 674; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 656 to 674; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 656 to 674.
  • such an amino acid sequence of the invention may be such that CDRl is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 232 to 250; and/or CDR2 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 444 to 462; and/or CDR3 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 656 to 674.
  • amino acid sequence of the invention essentially consists of 4 framework regions (FRl to FR4, respectively) and 3 complementarity determining regions (CDRl to CDR3, respectively), the amino acid sequence of the invention is preferably such that:
  • CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 250; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 232 to 250; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 232 to 250; and
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 444 to 462; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 444 to 462; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 444 to 462; and - CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 656 to 674; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 656 to 674; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 656 to 674; or any suitable fragment of such an amino acid sequence
  • such an amino acid sequence of the invention may be such that CDRl is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 232 to 250; and CDR2 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 444 to 462; and CDR3 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 656 to 674.
  • amino acid sequences are preferably such that they can specifically bind (as defined herein) to ADAM8; and more in particular bind to ADAM8 with an affinity (suitably measured and/or expressed as a K D -value (actual or apparent), a K A -value (actual or apparent), a kon-rate and/or a k o ff-rate, or alternatively as an IC50 value, as further described herein) that is as defined herein.
  • the invention relates to an amino acid sequence that essentially consists of 4 framework regions (FRl to FR4, respectively) and 3 complementarity determining regions (CDRl to CDR3, respectively), in which the CDR sequences of said amino acid sequence have at least 70% amino acid identity, preferably at least 80% amino acid identity, more preferably at least 90% amino acid identity, such as 95% amino acid identity or more or even essentially 100% amino acid identity with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 868 to 886.
  • This degree of amino acid identity can for example be determined by determining the degree of amino acid identity (in a manner described herein) between said amino acid sequence and one or more of the sequences of SEQ ID NO's: 868 to 886, in which the amino acid residues that form the framework regions are disregarded.
  • Such amino acid sequences of the invention can be as further described herein.
  • the invention relates to an amino acid sequence directed against ADAM9 (such as aNanobody of the invention, as further described herein), that comprises one or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; d) the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 4
  • an amino acid sequence of the invention contains one or more amino acid sequences according to b) and/or c): i) any amino acid substitution in such an amino acid sequence according to b) and/or c) is preferably, and compared to the corresponding amino acid sequence according to a), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to b) and/or c) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to a); and/or iii) the amino acid sequence according to b) and/or c) may be an amino acid sequence that is derived from an amino acid sequence according to a) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • an amino acid sequence of the invention contains one or more amino acid sequences according to e) and/or f): i) any amino acid substitution in such an amino acid sequence according to e) and/or f) is preferably, and compared to the corresponding amino acid sequence according to d), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to e) and/or f) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to d); and/or iii) the amino acid sequence according to e) and/or f) may be an amino acid sequence that is derived from an amino acid sequence according to d) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • an amino acid sequence of the invention contains one or more amino acid sequences according to h) and/or i): i) any amino acid substitution in such an amino acid sequence according to h) and/or i) is preferably, and compared to the corresponding amino acid sequence according to g), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to h) and/or i) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to g); and/or iii) the amino acid sequence according to h) and/or i) may be an amino acid sequence that is derived from an amino acid sequence according to g) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • amino acid sequences of the invention that comprise one or more amino acid sequences according to b), c), e), f), h) or i), respectively.
  • the amino acid sequence preferably comprises one or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; b) the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; and c) the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027; or any suitable combination thereof.
  • the invention relates to an amino acid sequence directed against ADAM9, that comprises two or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; d) the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; e) amino acid sequences that have at least 80%
  • the amino acid sequence preferably comprises two or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; b) the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; and c) the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027; such that, (i) when the first stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987, the second stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007 or of SEQ ID NO's: 675 to 695 and/or 1024 to 1027; (ii) when the first stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO'
  • the at least two stretches of amino acid residues again preferably form part of the antigen binding site for binding against ADAM9.
  • the invention relates to an amino acid sequence directed against ADAM9, that comprises three or more stretches of amino acid residues, in which the first stretch of amino acid residues is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; the second stretch of amino acid residues is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's:
  • the first stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; the second stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; and the third stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027.
  • the at least three stretches of amino acid residues forms part of the antigen binding site for binding against ADAM9.
  • the CDR sequences have at least 70% amino acid identity, preferably at least 80% amino acid identity, more preferably at least 90% amino acid identity, such as 95% amino acid identity or more or even essentially 100% amino acid identity with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 887 to 907 and 1044 to 1047.
  • This degree of amino acid identity can for example be determined by determining the degree of amino acid identity (in a manner described herein) between said amino acid sequence and one or more of the sequences of SEQ ID NO's: 887 to 907 and 1044 to 1047, in which the amino acid residues that form the framework regions are disregarded.
  • amino acid sequences of the invention can be as further described herein.
  • amino acid sequences are preferably such that they can specifically bind (as defined herein) to ADAM9; and more in particular bind to ADAM9 with an affinity (suitably measured and/or expressed as a K ⁇ -value (actual or apparent), a KA-value (actual or apparent), a k on -rate and/or a k off -rate, or alternatively as an IC 50 value, as further described herein) that is as defined herein.
  • amino acid sequence of the invention essentially consists of 4 framework regions (FRl to FR4, respectively) and 3 complementarity determining regions (CDRl to CDR3, respectively), the amino acid sequence of the invention is preferably such that:
  • CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; and/or
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; and/or
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027.
  • such an amino acid sequence of the invention may be such that CDRl is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; and/or CDR2 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; and/or CDR3 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027.
  • the amino acid sequence of the invention essentially consists of 4 framework regions (FRl to FR4, respectively) and 3 complementarity determining regions (CDRl to CDR3, respectively)
  • the amino acid sequence of the invention is preferably such that:
  • CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; and
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; and
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027; or any suitable fragment of such an amino acid sequence
  • such an amino acid sequence of the invention may be such that CDRl is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; and CDR2 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; and CDR3 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027.
  • amino acid sequences are preferably such that they can specifically bind (as defined herein) to ADAM9; and more in particular bind to ADAM9 with an affinity (suitably measured and/or expressed as a K ⁇ -value (actual or apparent), a KA-value (actual or apparent), a k on -rate and/or a k off -rate, or alternatively as an IC 5O value, as further described herein) that is as defined herein.
  • the invention relates to an amino acid sequence that essentially consists of 4 framework regions (FRl to FR4, respectively) and 3 complementarity determining regions (CDRl to CDR3, respectively), in which the CDR sequences of said amino acid sequence have at least 70% amino acid identity, preferably at least 80% amino acid identity, more preferably at least 90% amino acid identity, such as 95% amino acid identity or more or even essentially 100% amino acid identity with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 887 to 907 and 1044 to 1047.
  • This degree of amino acid identity can for example be determined by determining the degree of amino acid identity (in a manner described herein) between said amino acid sequence and one or more of the sequences of SEQ ID NO's: 887 to 907 and 1044 to 1047, in which the amino acid residues that form the framework regions are disregarded.
  • Such amino acid sequences of the invention can be as further described herein.
  • the invention relates to an amino acid sequence directed against ADAMlO (such as aNanobody of the invention, as further described herein), that comprises one or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 272 to 295; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 272 to 295; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 272 to 295; d) the amino acid sequences of SEQ ID NO's: 484 to 507; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 484 to 507; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequence
  • an amino acid sequence of the invention contains one or more amino acid sequences according to b) and/or c): i) any amino acid substitution in such an amino acid sequence according to b) and/or c) is preferably, and compared to the corresponding amino acid sequence according to a), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to b) and/or c) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to a); and/or iii) the amino acid sequence according to b) and/or c) may be an amino acid sequence that is derived from an amino acid sequence according to a) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • an amino acid sequence of the invention contains one or more amino acid sequences according to e) and/or f): i) any amino acid substitution in such an amino acid sequence according to e) and/or f) is preferably, and compared to the corresponding amino acid sequence according to d), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to e) and/or f) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to d); and/or iii) the amino acid sequence according to e) and/or f) may be an amino acid sequence that is derived from an amino acid sequence according to d) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • an amino acid sequence of the invention contains one or more amino acid sequences according to h) and/or i): i) any amino acid substitution in such an amino acid sequence according to h) and/or i) is preferably, and compared to the corresponding amino acid sequence according to g), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to h) and/or i) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to g); and/or iii) the amino acid sequence according to h) and/or i) may be an amino acid sequence that is derived from an amino acid sequence according to g) by means of affinity maturation using one or more techniques of affinity maturation known per se. It should be understood that the last preceding paragraphs also generally apply to any amino acid sequences of the invention that comprise one or more amino acid sequences according to b), c), e), f), h
  • the amino acid sequence preferably comprises one or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 272 to 295; b) the amino acid sequences of SEQ ID NO's: 484 to 507; and c) the amino acid sequences of SEQ ID NO's: 696 to 719; or any suitable combination thereof.
  • At least one of said stretches of amino acid residues forms part of the antigen binding site for binding against ADAMlO.
  • the invention relates to an amino acid sequence directed against ADAMlO, that comprises two or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 272 to 295; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 272 to 295; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 272 to 295; d) the amino acid sequences of SEQ ID NO's: 484 to 507; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 484 to 507; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 484 to 507; g
  • the amino acid sequence preferably comprises two or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 272 to 295; b) the amino acid sequences of SEQ ID NO's: 484 to 507; and c) the amino acid sequences of SEQ ID NO's: 696 to 719; such that, (i) when the first stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 272 to 295, the second stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 484 to 507 or of SEQ ID NO's: 696 to 719; (ii) when the first stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 484 to 507, the second stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 272 to 295 or of SEQ ID
  • the invention relates to an amino acid sequence directed against ADAMlO, that comprises three or more stretches of amino acid residues, in which the first stretch of amino acid residues is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 272 to 295; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 272 to 295; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 272 to 295; the second stretch of amino acid residues is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 484 to 507; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 484 to 507; f) amino acid sequences that have 3, 2, or 1 amino
  • the first stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 272 to 295; the second stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 484 to 507; and the third stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 696 to 719.
  • the at least three stretches of amino acid residues forms part of the antigen binding site for binding against ADAMlO.
  • the CDR sequences have at least 70% amino acid identity, preferably at least 80% amino acid identity, more preferably at least 90% amino acid identity, such as 95% amino acid identity or more or even essentially 100% amino acid identity with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 908 to 931.
  • This degree of amino acid identity can for example be determined by determining the degree of amino acid identity (in a manner described herein) between said amino acid sequence and one or more of the sequences of SEQ ID NO's: 908 to 931, in which the amino acid residues that form the framework regions are disregarded.
  • amino acid sequences of the invention can be as further described herein.
  • amino acid sequences are preferably such that they can specifically bind (as defined herein) to ADAMlO; and more in particular bind to ADAMlO with an affinity (suitably measured and/or expressed as a K D -value (actual or apparent), a K A -value (actual or apparent), a kon-rate and/or a k o ff-rate, or alternatively as an IC50 value, as further described herein) that is as defined herein.
  • amino acid sequence of the invention essentially consists of 4 framework regions (FRl to FR4, respectively) and 3 complementarity determining regions (CDRl to CDR3, respectively), the amino acid sequence of the invention is preferably such that:
  • CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 272 to 295; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 272 to 295; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 272 to 295; and/or
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 484 to 507; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 484 to 507; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 484 to 507; and/or
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 696 to 719; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 696 to 719; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 696 to 719.
  • such an amino acid sequence of the invention may be such that CDRl is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 272 to 295; and/or CDR2 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 484 to 507; and/or CDR3 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 696 to 719.
  • the amino acid sequence of the invention is preferably such that: - CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 272 to 295; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 272 to 295; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 272 to 295; and
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 484 to 507; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 484 to 507; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 484 to 507; and
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 696 to 719; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 696 to 719; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 696 to 719; or any suitable fragment of such an amino acid sequence
  • such an amino acid sequence of the invention may be such that CDRl is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 272 to 295; and CDR2 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 484 to 507; and CDR3 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 696 to 719.
  • amino acid sequences are preferably such that they can specifically bind (as defined herein) to ADAMlO; and more in particular bind to ADAMlO with an affinity (suitably measured and/or expressed as a K D -value (actual or apparent), a K A -value (actual or apparent), a kon-rate and/or a k o ff-rate, or alternatively as an IC50 value, as further described herein) that is as defined herein.
  • the invention relates to an amino acid sequence that essentially consists of 4 framework regions (FRl to FR4, respectively) and 3 complementarity determining regions (CDRl to CDR3, respectively), in which the CDR sequences of said amino acid sequence have at least 70% amino acid identity, preferably at least 80% amino acid identity, more preferably at least 90% amino acid identity, such as 95% amino acid identity or more or even essentially 100% amino acid identity with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 908 to 931.
  • This degree of amino acid identity can for example be determined by determining the degree of amino acid identity (in a manner described herein) between said amino acid sequence and one or more of the sequences of SEQ ID NO's: 908 to 931, in which the amino acid residues that form the framework regions are disregarded.
  • Such amino acid sequences of the invention can be as further described herein.
  • the invention relates to an amino acid sequence directed against ADAM 17 (such as a Nanobody of the invention, as further described herein), that comprises one or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; d) the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 508
  • an amino acid sequence of the invention contains one or more amino acid sequences according to b) and/or c): i) any amino acid substitution in such an amino acid sequence according to b) and/or c) is preferably, and compared to the corresponding amino acid sequence according to a), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to b) and/or c) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to a); and/or iii) the amino acid sequence according to b) and/or c) may be an amino acid sequence that is derived from an amino acid sequence according to a) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • an amino acid sequence of the invention contains one or more amino acid sequences according to e) and/or f): i) any amino acid substitution in such an amino acid sequence according to e) and/or f) is preferably, and compared to the corresponding amino acid sequence according to d), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to e) and/or f) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to d); and/or iii) the amino acid sequence according to e) and/or f) may be an amino acid sequence that is derived from an amino acid sequence according to d) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • an amino acid sequence of the invention contains one or more amino acid sequences according to h) and/or i): i) any amino acid substitution in such an amino acid sequence according to h) and/or i) is preferably, and compared to the corresponding amino acid sequence according to g), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to h) and/or i) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to g); and/or iii) the amino acid sequence according to h) and/or i) may be an amino acid sequence that is derived from an amino acid sequence according to g) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • amino acid sequences of the invention that comprise one or more amino acid sequences according to b), c), e), f), h) or i), respectively.
  • the amino acid sequence preferably comprises one or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; b) the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; and c) the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032; or any suitable combination thereof.
  • At least one of said stretches of amino acid residues forms part of the antigen binding site for binding against ADAM 17.
  • the invention relates to an amino acid sequence directed against ADAM 17, that comprises two or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; d) the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; f) amino acid sequence directed against ADAM 17, that comprises two or more
  • the amino acid sequence preferably comprises two or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; b) the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; and c) the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032; such that, (i) when the first stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992, the second stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012 or of SEQ ID NO's: 720 to 736 and/or 1028 to 1032; (ii) when the first stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ
  • the invention relates to an amino acid sequence directed against ADAM 17, that comprises three or more stretches of amino acid residues, in which the first stretch of amino acid residues is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; the second stretch of amino acid residues is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid
  • the first stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; the second stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; and the third stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032.
  • the at least three stretches of amino acid residues forms part of the antigen binding site for binding against ADAM 17.
  • the CDR sequences have at least 70% amino acid identity, preferably at least 80% amino acid identity, more preferably at least 90% amino acid identity, such as 95% amino acid identity or more or even essentially 100% amino acid identity with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 932 to 948 and 1048 to 1052 .
  • This degree of amino acid identity can for example be determined by determining the degree of amino acid identity (in a manner described herein) between said amino acid sequence and one or more of the sequences of SEQ ID NO's: 932 to 948 and 1048 to 1052 , in which the amino acid residues that form the framework regions are disregarded. Also, such amino acid sequences of the invention can be as further described herein.
  • amino acid sequences are preferably such that they can specifically bind (as defined herein) to ADAM 17; and more in particular bind to ADAM 17 with an affinity (suitably measured and/or expressed as a K D -value (actual or apparent), a K A -value (actual or apparent), a kon-rate and/or a k o ff-rate, or alternatively as an IC50 value, as further described herein) that is as defined herein.
  • amino acid sequence of the invention essentially consists of 4 framework regions (FRl to FR4, respectively) and 3 complementarity determining regions (CDRl to CDR3, respectively), the amino acid sequence of the invention is preferably such that:
  • CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; and/or
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; and/or
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032.
  • such an amino acid sequence of the invention may be such that CDRl is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; and/or CDR2 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; and/or CDR3 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032.
  • amino acid sequence of the invention essentially consists of 4 framework regions (FRl to FR4, respectively) and 3 complementarity determining regions (CDRl to CDR3, respectively), the amino acid sequence of the invention is preferably such that:
  • CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; and
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; and - CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino
  • such an amino acid sequence of the invention may be such that CDRl is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; and CDR2 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; and CDR3 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032.
  • CDRl is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992
  • CDR2 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012
  • CDR3 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032.
  • amino acid sequences are preferably such that they can specifically bind (as defined herein) to ADAM 17; and more in particular bind to ADAM 17 with an affinity (suitably measured and/or expressed as a K ⁇ -value (actual or apparent), a KA-value (actual or apparent), a kon-rate and/or a k o ff-rate, or alternatively as an IC50 value, as further described herein) that is as defined herein.
  • the invention relates to an amino acid sequence that essentially consists of 4 framework regions (FRl to FR4, respectively) and 3 complementarity determining regions (CDRl to CDR3, respectively), in which the CDR sequences of said amino acid sequence have at least 70% amino acid identity, preferably at least 80% amino acid identity, more preferably at least 90% amino acid identity, such as 95% amino acid identity or more or even essentially 100% amino acid identity with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 932 to 948 and 1048 to 1052 .
  • This degree of amino acid identity can for example be determined by determining the degree of amino acid identity (in a manner described herein) between said amino acid sequence and one or more of the sequences of SEQ ID NO's: 932 to 948 and 1048 to 1052, in which the amino acid residues that form the framework regions are disregarded.
  • Such amino acid sequences of the invention can be as further described herein.
  • the invention relates to an amino acid sequence directed against ADAMTS5 (such as a Nanobody of the invention, as further described herein), that comprises one or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; d) the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013; f) amino acid sequences that have 3, 2,
  • an amino acid sequence of the invention contains one or more amino acid sequences according to b) and/or c): i) any amino acid substitution in such an amino acid sequence according to b) and/or c) is preferably, and compared to the corresponding amino acid sequence according to a), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to b) and/or c) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to a); and/or iii) the amino acid sequence according to b) and/or c) may be an amino acid sequence that is derived from an amino acid sequence according to a) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • an amino acid sequence of the invention contains one or more amino acid sequences according to e) and/or f): i) any amino acid substitution in such an amino acid sequence according to e) and/or f) is preferably, and compared to the corresponding amino acid sequence according to d), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to e) and/or f) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to d); and/or iii) the amino acid sequence according to e) and/or f) may be an amino acid sequence that is derived from an amino acid sequence according to d) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • an amino acid sequence of the invention contains one or more amino acid sequences according to h) and/or i): i) any amino acid substitution in such an amino acid sequence according to h) and/or i) is preferably, and compared to the corresponding amino acid sequence according to g), a conservative amino acid substitution, (as defined herein); and/or ii) the amino acid sequence according to h) and/or i) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding amino acid sequence according to g); and/or iii) the amino acid sequence according to h) and/or i) may be an amino acid sequence that is derived from an amino acid sequence according to g) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • amino acid sequences of the invention that comprise one or more amino acid sequences according to b), c), e), f), h) or i), respectively.
  • the amino acid sequence preferably comprises one or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; b) the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013; and c) the amino acid sequences of SEQ ID NO's: 737 to 761 or 1033; or any suitable combination thereof.
  • the invention relates to an amino acid sequence directed against ADAMTS5, that comprises two or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; d) the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID
  • the amino acid sequence preferably comprises two or more stretches of amino acid residues chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; b) the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013; and c) the amino acid sequences of SEQ ID NO's: 737 to 761 or 1033; such that, (i) when the first stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 313 to 337 or 993, the second stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013 or of SEQ ID NO's: 737 to 761 or 1033; (ii) when the first stretch of amino acid residues corresponds to one of the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013, the second stretch of amino acid residues corresponds to one of the amino acid residues chosen from
  • the at least two stretches of amino acid residues again preferably form part of the antigen binding site for binding against ADAMTS5.
  • the invention relates to an amino acid sequence directed against ADAMTS5, that comprises three or more stretches of amino acid residues, in which the first stretch of amino acid residues is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; the second stretch of amino acid residues is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013
  • the first stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 313 to 337 or 993;
  • the second stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013;
  • the third stretch of amino acid residues is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 737 to 761 or 1033.
  • the at least three stretches of amino acid residues forms part of the antigen binding site for binding against ADAMTS5.
  • the CDR sequences have at least 70% amino acid identity, preferably at least 80% amino acid identity, more preferably at least 90% amino acid identity, such as 95% amino acid identity or more or even essentially 100% amino acid identity with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 949 to 974 and 1053 .
  • This degree of amino acid identity can for example be determined by determining the degree of amino acid identity (in a manner described herein) between said amino acid sequence and one or more of the sequences of SEQ ID NO's: 949 to 974 and 1053 , in which the amino acid residues that form the framework regions are disregarded.
  • amino acid sequences of the invention can be as further described herein.
  • amino acid sequences are preferably such that they can specifically bind (as defined herein) to ADAMTS5; and more in particular bind to ADAMTS5 with an affinity (suitably measured and/or expressed as a K ⁇ -value (actual or apparent), a KA-value (actual or apparent), a k on -rate and/or a k off -rate, or alternatively as an IC 50 value, as further described herein) that is as defined herein.
  • amino acid sequence of the invention essentially consists of 4 framework regions (FRl to FR4, respectively) and 3 complementarity determining regions (CDRl to CDR3, respectively), the amino acid sequence of the invention is preferably such that:
  • CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; and/or
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013; and/or
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 737 to 761 or 1033; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 737 to 761 or 1033; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 737 to 761 or 1033.
  • such an amino acid sequence of the invention may be such that CDRl is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; and/or CDR2 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013; and/or CDR3 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 737 to 761 or 1033.
  • amino acid sequence of the invention essentially consists of 4 framework regions (FRl to FR4, respectively) and 3 complementarity determining regions (CDRl to CDR3, respectively), the amino acid sequence of the invention is preferably such that:
  • CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; and
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013; and
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 737 to 761 or 1033; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 737 to 761 or 1033; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 737 to 761 or 1033; or any suitable fragment of such an amino acid sequence
  • such an amino acid sequence of the invention may be such that CDRl is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 313 to 337 or 993; and CDR2 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 525 to 549 or 1013; and CDR3 is chosen from the group consisting of the amino acid sequences of SEQ ID NO's: 737 to 761 or 1033.
  • amino acid sequences are preferably such that they can specifically bind (as defined herein) to ADAMTS5; and more in particular bind to ADAMTS5 with an affinity (suitably measured and/or expressed as a K ⁇ -value (actual or apparent), a KA-value (actual or apparent), a k ⁇ -rate and/or a k off -rate, or alternatively as an IC 50 value, as further described herein) that is as defined herein.
  • the invention relates to an amino acid sequence that essentially consists of 4 framework regions (FRl to FR4, respectively) and 3 complementarity determining regions (CDRl to CDR3, respectively), in which the CDR sequences of said amino acid sequence have at least 70% amino acid identity, preferably at least 80% amino acid identity, more preferably at least 90% amino acid identity, such as 95% amino acid identity or more or even essentially 100% amino acid identity with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 949 to 974 and 1053 .
  • This degree of amino acid identity can for example be determined by determining the degree of amino acid identity (in a manner described herein) between said amino acid sequence and one or more of the sequences of SEQ ID NO's: 949 to 974 and 1053, in which the amino acid residues that form the framework regions are disregarded.
  • Such amino acid sequences of the invention can be as further described herein.
  • the framework sequences may be any suitable framework sequences, and examples of suitable framework sequences will be clear to the skilled person, for example on the basis the standard handbooks and the further disclosure and prior art mentioned herein.
  • the framework sequences are preferably (a suitable combination of) immunoglobulin framework sequences or framework sequences that have been derived from immunoglobulin framework sequences (for example, by humanization or camelization).
  • the framework sequences may be framework sequences derived from a light chain variable domain (e.g. a VL-sequence) and/or from a heavy chain variable domain (e.g. a VH- sequence).
  • the framework sequences are either framework sequences that have been derived from a V HH - sec l uence ( m which said framework sequences may optionally have been partially or fully humanized) or are conventional VH sequences that have been camelized (as defined herein).
  • the framework sequences are preferably such that the amino acid sequence of the invention is a domain antibody (or an amino acid sequence that is suitable for use as a domain antibody); is a single domain antibody (or an amino acid sequence that is suitable for use as a single domain antibody); is a "dAb” (or an amino acid sequence that is suitable for use as a dAb); or is a NanobodyTM (including but not limited to VHH sequence).
  • suitable framework sequences will be clear to the skilled person, for example on the basis the standard handbooks and the further disclosure and prior art mentioned herein.
  • the framework sequences present in the amino acid sequences of the invention may contain one or more of Hallmark residues (as defined herein), such that the amino acid sequence of the invention is a NanobodyTM.
  • Hallmark residues as defined herein
  • the amino acid sequence of the invention is a NanobodyTM.
  • fragments that contain one or more CDR sequences, suitably flanked by and/or linked via one or more framework sequences (for example, in the same order as these CDR' s and framework sequences may occur in the full-sized immunoglobulin sequence from which the fragment has been derived).
  • Such fragments may also again be such that they comprise or can form an immunoglobulin fold, or alternatively be such that they do not comprise or cannot form an immunoglobulin fold.
  • such a fragment comprises a single CDR sequence as described herein (and in particular a CDR3 sequence), that is flanked on each side by (part of) a framework sequence (and in particular, part of the framework sequence(s) that, in the immunoglobulin sequence from which the fragment is derived, are adjacent to said CDR sequence.
  • a CDR3 sequence may be preceded by (part of) a FR3 sequence and followed by (part of) a FR4 sequence).
  • Such a fragment may also contain a disulphide bridge, and in particular a disulphide bridge that links the two framework regions that precede and follow the CDR sequence, respectively (for the purpose of forming such a disulphide bridge, cysteine residues that naturally occur in said framework regions may be used, or alternatively cysteine residues may be synthetically added to or introduced into said framework regions).
  • a disulphide bridge for the purpose of forming such a disulphide bridge, cysteine residues that naturally occur in said framework regions may be used, or alternatively cysteine residues may be synthetically added to or introduced into said framework regions.
  • the invention relates to a compound or construct, and in particular a protein or polypeptide (also referred to herein as a ""compound of the invention' or “polypeptide of the invention", respectively) that comprises or essentially consists of one or more amino acid sequences of the invention (or suitable fragments thereof), and optionally further comprises one or more other groups, residues, moieties or binding units.
  • a protein or polypeptide also referred to herein as a ""compound of the invention' or “polypeptide of the invention"
  • polypeptide of the invention comprises or essentially consists of one or more amino acid sequences of the invention (or suitable fragments thereof)
  • such further groups, residues, moieties, binding units or amino acid sequences may or may not provide further functionality to the amino acid sequence of the invention (and/or to the compound or construct in which it is present) and may or may not modify the properties of the amino acid sequence of the invention.
  • such further groups, residues, moieties or binding units may be one or more additional amino acid sequences, such that the compound or construct is a (fusion) protein or (fusion) polypeptide.
  • said one or more other groups, residues, moieties or binding units are immunoglobulin sequences.
  • said one or more other groups, residues, moieties or binding units are chosen from the group consisting of domain antibodies, amino acid sequences that are suitable for use as a domain antibody, single domain antibodies, amino acid sequences that are suitable for use as a single domain antibody, "dAb's", amino acid sequences that are suitable for use as a dAb, or Nanobodies.
  • such groups, residues, moieties or binding units may for example be chemical groups, residues, moieties, which may or may not by themselves be biologically and/or pharmacologically active.
  • such groups may be linked to the one or more amino acid sequences of the invention so as to provide a "derivative" of an amino acid sequence or polypeptide of the invention, as further described herein.
  • compounds or constructs that comprises or essentially consists of one or more derivatives as described herein, and optionally further comprises one or more other groups, residues, moieties or binding units, optionally linked via one or more linkers.
  • said one or more other groups, residues, moieties or binding units are amino acid sequences.
  • the one or more amino acid sequences of the invention and the one or more groups, residues, moieties or binding units may be linked directly to each other and/or via one or more suitable linkers or spacers.
  • the linkers may also be amino acid sequences, so that the resulting compound or construct is a fusion (protein) or fusion (polypeptide).
  • the amino acid sequences of the invention can be used as "building blocks" to form polypeptides of the invention, i.e.
  • the compounds or polypeptides of the invention can generally be prepared by a method which comprises at least one step of suitably linking the one or more amino acid sequences of the invention to the one or more further groups, residues, moieties or binding units, optionally via the one or more suitable linkers, so as to provide the compound or polypeptide of the invention.
  • Polypeptides of the invention can also be prepared by a method which generally comprises at least the steps of providing a nucleic acid that encodes a polypeptide of the invention, expressing said nucleic acid in a suitable manner, and recovering the expressed polypeptide of the invention. Such methods can be performed in a manner known per se, which will be clear to the skilled person, for example on the basis of the methods and techniques further described herein.
  • a multispecific polypeptide of the invention may at least comprise at least one amino acid sequence (such as a Nanobody) against an ADAM proteinase and at least one binding unit against TNF (such as the Nanobodies described in the international applications WO 06/122786 and WO 04/041862 of Ablynx N.V. or the dAb's described in WO 03/002609 or WO 04/003019).
  • a multispecific polypeptide may for example be used in the prevention and treatment of inflammation and other diseases and disorders associated with TNF and the TNF pathways. Reference is made to WO 06/122786, WO 04/041862, WO 03/002609 or WO 04/003019 and WO 06/038027.
  • Another multispecific polypeptide of the invention may at least comprise at least one amino acid sequence (such as a Nanobody) against an ADAM proteinase (which amino acid sequence is preferably not inactivating) and at least one binding unit against a receptor so as to induce shedding and inactivation of said receptor.
  • a multispecific polypeptide of the invention (as defined herein) may at least comprise at least one amino acid sequence (such as a Nanobody) against an ADAM proteinase (which amino acid sequence is preferably not inactivating) and at least one binding unit against a TNF receptor (e.g. TNFRl or TNFR2) so as to induce shedding and inactivation of said TNF receptor (such as the dAb's described in WO 06/038027).
  • Such a multispecific polypeptide may for example be used in the prevention and treatment of inflammation and other diseases and disorders associated with TNF and the TNF pathways.
  • Another multispecific polypeptide of the invention may at least comprise at least one amino acid sequence (such as a Nanobody) against an ADAM proteinase (which amino acid sequence is preferably not inactivating) and at least one binding unit against APP so as to cleave or solubilize APP aggregates.
  • at least one amino acid sequence such as a Nanobody
  • ADAM proteinase which amino acid sequence is preferably not inactivating
  • binding unit against APP so as to cleave or solubilize APP aggregates.
  • Another multispecific polypeptide of the invention may at least comprise at least two amino acid sequence (such as two Nanobodies) against different ADAM proteinase so as to provide an increased and potentially synergistic modulating (and preferably inhibiting) effect on the ADAM proteinases.
  • at least two amino acid sequence such as two Nanobodies
  • a compound of the invention or a polypeptide of the invention may have an increased half-life, compared to the corresponding amino acid sequence of the invention.
  • Some preferred, but non-limiting examples of such compounds and polypeptides will become clear to the skilled person based on the further disclosure herein, and for example comprise amino acid sequences or polypeptides of the invention that have been chemically modified to increase the half-life thereof (for example, by means of pegylation); amino acid sequences of the invention that comprise at least one additional binding site for binding to a serum protein (such as serum albumin); or polypeptides of the invention that comprise at least one amino acid sequence of the invention that is linked to at least one moiety (and in particular at least one amino acid sequence) that increases the half- life of the amino acid sequence of the invention.
  • polypeptides of the invention that comprise such half-life extending moieties or amino acid sequences will become clear to the skilled person based on the further disclosure herein; and for example include, without limitation, polypeptides in which the one or more amino acid sequences of the invention are suitable linked to one or more serum proteins or fragments thereof (such as (human) serum albumin or suitable fragments thereof) or to one or more binding units that can bind to serum proteins (such as, for example, domain antibodies, amino acid sequences that are suitable for use as a domain antibody, single domain antibodies, amino acid sequences that are suitable for use as a single domain antibody, "dAb's", amino acid sequences that are suitable for use as a dAb, or Nanobodies that can bind to serum proteins such as serum albumin (such as human serum albumin), serum immunoglobulins such as IgG, or transferrin; reference is made to the further description and references mentioned herein); polypeptides in which an amino acid sequence of the invention is linked to an Fc portion (such as a human
  • the compounds or polypeptides of the invention with increased half-life preferably have a half-life that is at least 1.5 times, preferably at least 2 times, such as at least 5 times, for example at least 10 times or more than 20 times, greater than the half-life of the corresponding amino acid sequence of the invention per se.
  • the compounds or polypeptides of the invention with increased half-life may have a half-life that is increased with more than 1 hours, preferably more than 2 hours, more preferably more than 6 hours, such as more than 12 hours, or even more than 24, 48 or 72 hours, compared to the corresponding amino acid sequence of the invention per se.
  • such compounds or polypeptides of the invention have a serum half-life that is increased with more than 1 hours, preferably more than 2 hours, more preferably more than 6 hours, such as more than 12 hours, or even more than 24, 48 or 72 hours, compared to the corresponding amino acid sequence of the invention per se.
  • such compounds or polypeptides of the invention exhibit a serum half-life in human of at least about 12 hours, preferably at least 24 hours, more preferably at least 48 hours, even more preferably at least 72 hours or more.
  • compounds or polypeptides of the invention may have a half- life of at least 5 days (such as about 5 to 10 days), preferably at least 9 days (such as about 9 to 14 days), more preferably at least about 10 days (such as about 10 to 15 days), or at least about 11 days (such as about 11 to 16 days), more preferably at least about 12 days (such as about 12 to 18 days or more), or more than 14 days (such as about 14 to 19 days).
  • the invention relates to a nucleic acid that encodes an amino acid sequence of the invention or a polypeptide of the invention (or a suitable fragment thereof).
  • a nucleic acid will also be referred to herein as a ""nucleic acid of the invention' and may for example be in the form of a genetic construct, as further described herein.
  • the invention relates to a host or host cell that expresses (or that under suitable circumstances is capable of expressing) an amino acid sequence of the invention and/or a polypeptide of the invention; and/or that contains a nucleic acid of the invention.
  • the invention further relates to a product or composition containing or comprising at least one amino acid sequence of the invention, at least one polypeptide of the invention (or a suitable fragment thereof) and/or at least one nucleic acid of the invention, and optionally one or more further components of such compositions known per se, i.e. depending on the intended use of the composition.
  • a product or composition may for example be a pharmaceutical composition (as described herein), a veterinary composition or a product or composition for diagnostic use (as also described herein).
  • Some preferred but non-limiting examples of such products or compositions will become clear from the further description herein.
  • the invention further relates to methods for preparing or generating the amino acid sequences, polypeptides, nucleic acids, host cells, products and compositions described herein. Some preferred but non-limiting examples of such methods will become clear from the further description herein.
  • these methods may comprise the steps of: a) providing a set, collection or library of amino acid sequences; and b) screening said set, collection or library of amino acid sequences for amino acid sequences that can bind to and/or have affinity for a metalloproteinase from the ADAM family; and c) isolating the amino acid sequence(s) that can bind to and/or have affinity for a metalloproteinase from the ADAM family.
  • the set, collection or library of amino acid sequences may be any suitable set, collection or library of amino acid sequences.
  • the set, collection or library of amino acid sequences may be a set, collection or library of immunoglobulin sequences (as described herein), such as a na ⁇ ve set, collection or library of immunoglobulin sequences; a synthetic or semi-synthetic set, collection or library of immunoglobulin sequences; and/or a set, collection or library of immunoglobulin sequences that have been subjected to affinity maturation.
  • the set, collection or library of amino acid sequences may be a set, collection or library of heavy chain variable domains (such as V H domains or V HH domains) or of light chain variable domains.
  • the set, collection or library of amino acid sequences may be a set, collection or library of domain antibodies or single domain antibodies, or may be a set, collection or library of amino acid sequences that are capable of functioning as a domain antibody or single domain antibody.
  • the set, collection or library of amino acid sequences may be an immune set, collection or library of immunoglobulin sequences, for example derived from a mammal that has been suitably immunized with a metalloproteinase from the ADAM family or with a suitable antigenic determinant based thereon or derived therefrom, such as an antigenic part, fragment, region, domain, loop or other epitope thereof.
  • said antigenic determinant may be an extracellular part, region, domain, loop or other extracellular epitope(s).
  • the set, collection or library of amino acid sequences may be displayed on a phage, phagemid, ribosome or suitable micro-organism (such as yeast), such as to facilitate screening.
  • suitable methods, techniques and host organisms for displaying and screening (a set, collection or library of) amino acid sequences will be clear to the person skilled in the art, for example on the basis of the further disclosure herein. Reference is also made to the review by Hoogenboom in Nature Biotechnology, 23, 9, 1105-1116 (2005).
  • the method for generating amino acid sequences comprises at least the steps of: a) providing a collection or sample of cells expressing amino acid sequences; b) screening said collection or sample of cells for cells that express an amino acid sequence that can bind to and/or have affinity for a metalloproteinase from the ADAM family; and c) either (i) isolating said amino acid sequence; or (ii) isolating from said cell a nucleic acid sequence that encodes said amino acid sequence, followed by expressing said amino acid sequence.
  • the collection or sample of cells may for example be a collection or sample of B-cells.
  • the sample of cells may be derived from a mammal that has been suitably immunized with a metalloproteinase from the ADAM family or with a suitable antigenic determinant based thereon or derived therefrom, such as an antigenic part, fragment, region, domain, loop or other epitope thereof.
  • said antigenic determinant may be an extracellular part, region, domain, loop or other extracellular epitope(s).
  • step b) is preferably performed using a flow cytometry technique such as FACS.
  • FACS flow cytometry technique
  • the method for generating an amino acid sequence directed against a metalloproteinase from the ADAM family may comprise at least the steps of: a) providing a set, collection or library of nucleic acid sequences encoding amino acid sequences; b) screening said set, collection or library of nucleic acid sequences for nucleic acid sequences that encode an amino acid sequence that can bind to and/or has affinity for a metalloproteinase from the ADAM family; and c) isolating said nucleic acid sequence, followed by expressing said amino acid sequence.
  • the set, collection or library of nucleic acid sequences encoding amino acid sequences may for example be a set, collection or library of nucleic acid sequences encoding a na ⁇ ve set, collection or library of immunoglobulin sequences; a set, collection or library of nucleic acid sequences encoding a synthetic or semi-synthetic set, collection or library of immunoglobulin sequences; and/or a set, collection or library of nucleic acid sequences encoding a set, collection or library of immunoglobulin sequences that have been subjected to affinity maturation.
  • the set, collection or library of nucleic acid sequences may encode a set, collection or library of heavy chain variable domains (such as V H domains or V HH domains) or of light chain variable domains.
  • the set, collection or library of nucleic acid sequences may encode a set, collection or library of domain antibodies or single domain antibodies, or a set, collection or library of amino acid sequences that are capable of functioning as a domain antibody or single domain antibody.
  • the set, collection or library of amino acid sequences may be an immune set, collection or library of nucleic acid sequences, for example derived from a mammal that has been suitably immunized with a metalloproteinase from the ADAM family or with a suitable antigenic determinant based thereon or derived therefrom, such as an antigenic part, fragment, region, domain, loop or other epitope thereof.
  • said antigenic determinant may be an extracellular part, region, domain, loop or other extracellular epitope(s).
  • the set, collection or library of nucleic acid sequences may for example encode an immune set, collection or library of heavy chain variable domains or of light chain variable domains.
  • the set, collection or library of nucleotide sequences may encode a set, collection or library of VHH sequences.
  • the set, collection or library of nucleotide sequences may be displayed on a phage, phagemid, ribosome or suitable micro-organism (such as yeast), such as to facilitate screening.
  • suitable methods, techniques and host organisms for displaying and screening (a set, collection or library of) nucleotide sequences encoding amino acid sequences will be clear to the person skilled in the art, for example on the basis of the further disclosure herein. Reference is also made to the review by Hoogenboom in Nature Biotechnology, 23, 9, 1105-1116 (2005).
  • the invention also relates to amino acid sequences that are obtained by the above methods, or alternatively by a method that comprises the one of the above methods and in addition at least the steps of determining the nucleotide sequence or amino acid sequence of said immunoglobulin sequence; and of expressing or synthesizing said amino acid sequence in a manner known per se, such as by expression in a suitable host cell or host organism or by chemical synthesis.
  • one or more amino acid sequences of the invention may be suitably humanized (or alternatively camelized); and/or the amino acid sequence(s) thus obtained may be linked to each other or to one or more other suitable amino acid sequences (optionally via one or more suitable linkers) so as to provide a polypeptide of the invention.
  • nucleic acid sequence encoding an amino acid sequence of the invention may be suitably humanized (or alternatively camelized) and suitably expressed; and/or one or more nucleic acid sequences encoding an amino acid sequence of the invention may be linked to each other or to one or more nucleic acid sequences that encode other suitable amino acid sequences (optionally via nucleotide sequences that encode one or more suitable linkers), after which the nucleotide sequence thus obtained may be suitably expressed so as to provide a polypeptide of the invention.
  • the invention further relates to applications and uses of the amino acid sequences, polypeptides, nucleic acids, host cells, products and compositions described herein, as well as to methods for the prevention and/or treatment for diseases and disorders associated with a metalloproteinase from the ADAM family.
  • Nanobodies generally offer certain advantages (outlined herein) compared to "dAb's" or similar (single) domain antibodies or immunoglobulin sequences, which advantages are also provided by the Nanobodies of the invention.
  • advantages outlined herein
  • similar (single) domain antibodies or immunoglobulin sequences which advantages are also provided by the Nanobodies of the invention.
  • the more general aspects of the teaching below can also be applied (either directly or analogously) to other amino acid sequences of the invention.
  • immunoglobulin sequence whether used herein to refer to a heavy chain antibody or to a conventional 4-chain antibody - is used as a general term to include both the full-size antibody, the individual chains thereof, as well as all parts, domains or fragments thereof (including but not limited to antigen -binding domains or fragments such as V HH domains or V H /V L domains, respectively).
  • sequence as used herein (for example in terms like “immunoglobulin sequence”, “antibody sequence”, “variable domain sequence”, “VHH sequence” or “protein sequence”), should generally be understood to include both the relevant amino acid sequence as well as nucleic acids or nucleotide sequences encoding the same, unless the context requires a more limited interpretation.
  • nucleotide sequence as used herein also encompasses a nucleic acid molecule with said nucleotide sequence, so that the terms “nucleotide sequence” and “nucleic acid” should be considered equivalent and are used interchangeably herein; c) Unless indicated otherwise, all methods, steps, techniques and manipulations that are not specifically described in detail can be performed and have been performed in a manner known per se, as will be clear to the skilled person. Reference is for example again made to the standard handbooks and the general background art mentioned herein and to the further references cited therein; as well as to for example the following reviews Presta, Adv. Drug Deliv. Rev. 2006, 58 (5-6): 640-56; Levin and Weiss, MoI.
  • the percentage of "sequence identity" between a first nucleotide sequence and a second nucleotide sequence may be calculated by dividing [the number of nucleotides in the first nucleotide sequence that are identical to the nucleotides at the corresponding positions in the second nucleotide sequence] by [the total number of nucleotides in the first nucleotide sequence] and multiplying by [100%], in which each deletion, insertion, substitution or addition of a nucleotide in the second nucleotide sequence - compared to the first nucleotide sequence - is considered as a difference at a single nucleotide (position).
  • the degree of sequence identity between two or more nucleotide sequences may be calculated using a known computer algorithm for sequence alignment such as NCBI Blast v2.0, using standard settings.
  • the nucleotide sequence with the greatest number of nucleotides will be taken as the "first" nucleotide sequence, and the other nucleotide sequence will be taken as the "second" nucleotide sequence; f)
  • the percentage of "sequence identity" between a first amino acid sequence and a second amino acid sequence may be calculated by dividing [the number of amino acid residues in the first amino acid sequence that are identical to the amino acid residues at the corresponding positions in the second amino acid sequence] by [the total number of amino acid residues in the first amino acid sequence] and multiplying by [100%], in which each deletion, insertion, substitution or addition of an amino acid residue in the second amino acid sequence - compared to the first amino acid sequence - is
  • the degree of sequence identity between two amino acid sequences may be calculated using a known computer algorithm, such as those mentioned above for determining the degree of sequence identity for nucleotide sequences, again using standard settings.
  • amino acid sequence with the greatest number of amino acid residues will be taken as the "first" amino acid sequence, and the other amino acid sequence will be taken as the "second" amino acid sequence.
  • amino acid substitutions which can generally be described as amino acid substitutions in which an amino acid residue is replaced with another amino acid residue of similar chemical structure and which has little or essentially no influence on the function, activity or other biological properties of the polypeptide.
  • Such conservative amino acid substitutions are well known in the art, for example from WO 04/037999, GB-A-3 357 768, WO 98/49185, WO 00/46383 and WO 01/09300; and (preferred) types and/or combinations of such substitutions may be selected on the basis of the pertinent teachings from WO 04/037999 as well as WO 98/49185 and from the further references cited therein.
  • Such conservative substitutions preferably are substitutions in which one amino acid within the following groups (a) - (e) is substituted by another amino acid residue within the same group: (a) small aliphatic, nonpolar or slightly polar residues: Ala, Ser, Thr, Pro and GIy; (b) polar, negatively charged residues and their (uncharged) amides: Asp, Asn, GIu and GIn; (c) polar, positively charged residues: His, Arg and Lys; (d) large aliphatic, nonpolar residues: Met, Leu, He, VaI and Cys; and (e) aromatic residues: Phe, Tyr and Trp.
  • Particularly preferred conservative substitutions are as follows: Ala into GIy or into Ser; Arg into Lys; Asn into GIn or into His; Asp into GIu; Cys into Ser; GIn into Asn; GIu into Asp; GIy into Ala or into Pro; His into Asn or into GIn; He into Leu or into VaI; Leu into He or into VaI; Lys into Arg, into GIn or into GIu; Met into Leu, into Tyr or into He; Phe into Met, into Leu or into Tyr; Ser into Thr; Thr into Ser; Trp into Tyr;
  • Any amino acid substitutions applied to the polypeptides described herein may also be based on the analysis of the frequencies of amino acid variations between homologous proteins of different species developed by Schulz et al., Principles of Protein Structure, Springer-Verlag, 1978, on the analyses of structure forming potentials developed by Chou and Fasman, Biochemistry 13: 211, 1974 and Adv. EnzymoL, 47: 45-149, 1978, and on the analysis of hydrophobicity patterns in proteins developed by Eisenberg et al., Proc. Natl. Acad. Sci. USA 81: 140-144, 1984; Kyte & Doolittle; J Molec. Biol.
  • amino acid difference refers to an insertion, deletion or substitution of a single amino acid residue on a position of the first sequence, compared to the second sequence; it being understood that two amino acid sequences can contain one, two or more such amino acid differences; i) When a nucleotide sequence or amino acid sequence is said to "comprise” another nucleotide sequence or amino acid sequence, respectively, or to "essentially consist of another nucleotide sequence or amino acid sequence, this may mean that the latter nucleotide sequence or amino acid sequence has been incorporated into the firstmentioned nucleotide sequence or amino acid sequence, respectively, but more usually this generally means that the firstmentioned nucleotide sequence or amino acid sequence comprises within its sequence a stretch of nucleotides or amino acid sequence
  • aNanobody of the invention when aNanobody of the invention is said to comprise a CDR sequence, this may mean that said CDR sequence has been incorporated into the Nanobody of the invention, but more usually this generally means that the Nanobody of the invention contains within its sequence a stretch of amino acid residues with the same amino acid sequence as said CDR sequence, irrespective of how said Nanobody of the invention has been generated or obtained.
  • the latter amino acid sequence when it has a specific biological or structural function, it preferably has essentially the same, a similar or an equivalent biological or structural function in the firstmentioned amino acid sequence (in other words, the firstmentioned amino acid sequence is preferably such that the latter sequence is capable of performing essentially the same, a similar or an equivalent biological or structural function).
  • the CDR sequence and framework are preferably capable, in said Nanobody, of functioning as a CDR sequence or framework sequence, respectively.
  • nucleotide sequence when a nucleotide sequence is said to comprise another nucleotide sequence, the firstmentioned nucleotide sequence is preferably such that, when it is expressed into an expression product (e.g. a polypeptide), the amino acid sequence encoded by the latter nucleotide sequence forms part of said expression product (in other words, that the latter nucleotide sequence is in the same reading frame as the firstmentioned, larger nucleotide sequence), j) A nucleic acid sequence or amino acid sequence is considered to be "(in) essentially isolated (form)" - for example, compared to its native biological source and/or the reaction medium or cultivation medium from which it has been obtained - when it has been separated from at least one other component with which it is usually associated in said source or medium, such as another nucleic acid, another protein/polypeptide, another biological component or macromolecule or at least one contaminant, impurity or minor component.
  • an expression product e.g. a polypeptide
  • a nucleic acid sequence or amino acid sequence is considered “essentially isolated” when it has been purified at least 2- fold, in particular at least 10-fold, more in particular at least 100-fold, and up to 1000-fold or more.
  • a nucleic acid sequence or amino acid sequence that is "in essentially isolated form” is preferably essentially homogeneous, as determined using a suitable technique, such as a suitable chromatographical technique, such as polyacrylamide-gel electrophoresis; k)
  • domain as used herein generally refers to a globular region of an amino acid sequence (such as an antibody chain, and in particular to a globular region of a heavy chain antibody), or to a polypeptide that essentially consists of such a globular region.
  • binding domain refers to such a domain that is directed against an antigenic determinant (as defined herein); 1)
  • antigenic determinant refers to the epitope on the antigen recognized by the antigen-binding molecule (such as a Nanobody or a polypeptide of the invention) and more in particular by the antigen-binding site of said molecule.
  • antigenic determinant' and “epitope” may also be used interchangeably herein, m)
  • An amino acid sequence such as a Nanobody, an antibody, a polypeptide of the invention, or generally an antigen binding protein or polypeptide or a fragment thereof
  • an amino acid sequence that can (specifically) bind to, that has affinity for and/or that has specificity for a specific antigenic determinant, epitope, antigen or protein (or for at least one part, fragment or epitope thereof) is said to be "against” or “directed against” said antigenic determinant, epitope, antigen or protein.
  • the term "specificity" refers to the number of different types of antigens or antigenic determinants to which a particular antigen-binding molecule or antigen-binding protein (such as a Nanobody or a polypeptide of the invention) molecule can bind.
  • the specificity of an antigen -binding protein can be determined based on affinity and/or avidity.
  • the affinity represented by the equilibrium constant for the dissociation of an antigen with an antigen-binding protein (K D ), is a measure for the binding strength between an antigenic determinant and an antigen -binding site on the antigen-binding protein: the lesser the value of the K D , the stronger the binding strength between an antigenic determinant and the antigen-binding molecule (alternatively, the affinity can also be expressed as the affinity constant (KA), which is 1/KD).
  • affinity can be determined in a manner known per se, depending on the specific antigen of interest.
  • Avidity is the measure of the strength of binding between an antigen -binding molecule (such as a Nanobody or polypeptide of the invention) and the pertinent antigen. Avidity is related to both the affinity between an antigenic determinant and its antigen binding site on the antigen -binding molecule and the number of pertinent binding sites present on the antigen-binding molecule.
  • antigen-binding proteins such as the amino acid sequences, Nanobodies and/or polypeptides of the invention
  • K D dissociation constant
  • 7 12 -S 11 preferably 10 " to 10 " moles/liter or less and more preferably 10 to 10 " moles/liter
  • K A association constant
  • any K D value greater than 10 mo I/liter (or any K A value lower than 10 M " ) liters/mol is generally considered to indicate non-specific binding.
  • a monovalent immunoglobulin sequence of the invention will bind to the desired antigen with an affinity less than 500 nM, preferably less than 200 nM, more preferably less than 10 nM, such as less than 500 pM.
  • Specific binding of an antigen -binding protein to an antigen or antigenic determinant can be determined in any suitable manner known per se, including, for example, Scatchard analysis and/or competitive binding assays, such as radioimmunoassays (RIA), enzyme immunoassays (EIA) and sandwich competition assays, and the different variants thereof known per se in the art; as well as the other techniques mentioned herein.
  • the dissociation constant may be the actual or apparent dissociation constant, as will be clear to the skilled person. Methods for determining the dissociation constant will be clear to the skilled person, and for example include the techniques mentioned herein.
  • the affinity denotes the strength or stability of a molecular interaction.
  • the affinity is commonly given as by the K D , or dissociation constant, which has units of mo I/liter (or M).
  • the affinity can also be expressed as an association constant, KA, which equals 1/KD and has units of (mol/liter) " (or M " ).
  • the K D for biological interactions which are considered meaningful are typically in the range of 10 "10 M (0.1 nM) to 10 "5 M (10000 nM). The stronger an interaction is, the lower is its KD.
  • the off-rate k o ff has units s " (where s is the SI unit notation of second).
  • the on-rate k on has units M -1 S "1 .
  • the on-rate may vary between IO 2 M -1 S "1 to about 10 7 M 4 S "1 , approaching the diffusion-limited association rate constant for bimolecular interactions.
  • the affinity of a molecular interaction between two molecules can be measured via different techniques known per se, such as the well known surface plasmon resonance
  • the measured KD may correspond to the apparent K D if the measuring process somehow influences the intrinsic binding affinity of the implied molecules for example by artefacts related to the coating on the biosensor of one molecule. Also, an apparent KD may be measured if one molecule contains more than one recognition sites for the other molecule. In such situation the measured affinity may be affected by the avidity of the interaction by the two molecules.
  • a reference molecule C that is known to bind to B and that is suitably labelled with a fluorophore or chromophore group or other chemical moiety, such as biotin for easy detection in an ELISA or FACS (Fluorescent activated cell sorting) or other format (the fluorophore for fluorescence detection, the chromophore for light absorption detection, the biotin for streptavidin-mediated ELISA detection).
  • the reference molecule C is kept at a fixed concentration and the concentration of A is varied for a given concentration or amount of B. As a result an IC 5 o value is obtained corresponding to the concentration of A at which the signal measured for C in absence of A is halved.
  • KDref the KD of the reference molecule
  • c ref the total concentration of the reference molecule
  • KD ⁇ IC50 KD ⁇ IC50.
  • the half-life of an amino acid sequence, compound or polypeptide of the invention can generally be defined as the time taken for the serum concentration of the amino acid sequence, compound or polypeptide to be reduced by 50%, in vivo, for example due to degradation of the sequence or compound and/or clearance or sequestration of the sequence or compound by natural mechanisms.
  • the in vivo half-life of an amino acid sequence, compound or polypeptide of the invention can be determined in any manner known per se, such as by pharmacokinetic analysis. Suitable techniques will be clear to the person skilled in the art, and may for example generally involve the steps of suitably administering to a warm-blooded animal (i.e.
  • a human or to another suitable mammal such as a mouse, rabbit, rat, pig, dog or a primate, for example monkeys from the genus Macaca (such as, and in particular, cynomolgus monkeys ⁇ Macaca fascicularis) and/or rhesus monkeys ⁇ Macaca mulatto)) and baboon (Papio ursinus)) a suitable dose of the amino acid sequence, compound or polypeptide of the invention; collecting blood samples or other samples from said animal; determining the level or concentration of the amino acid sequence, compound or polypeptide of the invention in said blood sample; and calculating, from (a plot of) the data thus obtained, the time until the level or concentration of the amino acid sequence, compound or polypeptide of the invention has been reduced by 50% compared to the initial level upon dosing.
  • Macaca such as, and in particular, cynomolgus monkeys ⁇ Macaca fascicularis) and/or rhesus monkeys
  • the half-life can be expressed using parameters such as the tl/2-alpha, tl/2-beta and the area under the curve (AUC).
  • an "increase in half-life” refers to an increase in any one of these parameters, such as any two of these parameters, or essentially all three these parameters.
  • increase in half-life or “increased half-life” in particular refers to an increase in the tl/2-beta, either with or without an increase in the tl/2-alpha and/or the AUC or both.
  • modulating or “to modulate” generally means either reducing or inhibiting the activity of, or alternatively increasing the activity of, a target or antigen, as measured using a suitable in vitro, cellular or in vivo assay.
  • modulating or “to modulate” may mean either reducing or inhibiting the activity of, or alternatively increasing a (relevant or intended) biological activity of, a target or antigen, as measured using a suitable in vitro, cellular or in vivo assay (which will usually depend on the target or antigen involved), by at least 1%, preferably at least 5%, such as at least 10% or at least 25%, for example by at least 50%, at least 60%, at least 70%, at least 80%, or 90% or more, compared to activity of the target or antigen in the same assay under the same conditions but without the presence of the construct of the invention.
  • moduleating may also involve effecting a change (which may either be an increase or a decrease) in affinity, avidity, specificity and/or selectivity of a target or antigen for one or more of its ligands, binding partners, partners for association into a homomultimeric or heteromultimeric form, or substrates; and/or effecting a change (which may either be an increase or a decrease) in the sensitivity of the target or antigen for one or more conditions in the medium or surroundings in which the target or antigen is present (such as pH, ion strength, the presence of co-factors, etc.), compared to the same conditions but without the presence of the construct of the invention.
  • this may again be determined in any suitable manner and/or using any suitable assay known per se, depending on the target or antigen involved.
  • Modulating may also mean effecting a change (i.e. an activity as an agonist, as an antagonist or as a reverse agonist, respectively, depending on the target or antigen and the desired biological or physiological effect) with respect to one or more biological or physiological mechanisms, effects, responses, functions, pathways or activities in which the target or antigen (or in which its substrate(s), ligand(s) or pathway(s) are involved, such as its signalling pathway or metabolic pathway and their associated biological or physiological effects) is involved.
  • a change i.e. an activity as an agonist, as an antagonist or as a reverse agonist, respectively, depending on the target or antigen and the desired biological or physiological effect
  • a change i.e. an activity as an agonist, as an antagonist or as a reverse agonist, respectively, depending on the target or antigen and the desired biological or physiological effect
  • a change i.e. an activity as an agonist, as an antagonist or as a reverse agonist, respectively, depending on the target or antigen and the desired biological or physiological effect
  • an action as an agonist or antagonist may be such that an intended biological or physiological activity is increased or decreased, respectively, by at least 1%, preferably at least 5%, such as at least 10% or at least 25%, for example by at least 50%, at least 60%, at least 70%, at least 80%, or 90% or more, compared to the biological or physiological activity in the same assay under the same conditions but without the presence of the construct of the invention.
  • Modulating may for example also involve allosteric modulation of the target or antigen; and/or reducing or inhibiting the binding of the target or antigen to one of its substrates or ligands and/or competing with a natural ligand, substrate for binding to the target or antigen. Modulating may also involve activating the target or antigen or the mechanism or pathway in which it is involved. Modulating may for example also involve effecting a change in respect of the folding or confirmation of the target or antigen, or in respect of the ability of the target or antigen to fold, to change its confirmation (for example, upon binding of a ligand), to associate with other (sub)units, or to disassociate. Modulating may for example also involve effecting a change in the ability of the target or antigen to transport other compounds or to serve as a channel for other compounds (such as ions).
  • the term "interaction site" on the target or antigen means a site, epitope, antigenic determinant, part, domain or stretch of amino acid residues on the target or antigen that is a site for binding to a ligand, receptor or other binding partner, a catalytic site, a cleavage site, a site for allosteric interaction, a site involved in multimerisation (such as homomerization or heterodimerization) of the target or antigen; or any other site, epitope, antigenic determinant, part, domain or stretch of amino acid residues on the target or antigen that is involved in a biological action or mechanism of the target or antigen.
  • an "interaction site” can be any site, epitope, antigenic determinant, part, domain or stretch of amino acid residues on the target or antigen to which an amino acid sequence or polypeptide of the invention can bind such that the target or antigen (and/or any pathway, interaction, signalling, biological mechanism or biological effect in which the target or antigen is involved) is modulated (as defined herein).
  • An amino acid sequence or polypeptide is said to be "specific for" a first target or antigen compared to a second target or antigen when is binds to the first antigen with an affinity (as described above, and suitably expressed as a K D value, K A value, K 0 ⁇ rate and/or K 0n rate) that is at least 10 times, such as at least 100 times, and preferably at least 1000 times, and up to 10.000 times or more better than the affinity with which said amino acid sequence or polypeptide binds to the second target or polypeptide.
  • an affinity as described above, and suitably expressed as a K D value, K A value, K 0 ⁇ rate and/or K 0n rate
  • the first antigen may bind to the target or antigen with a K D value that is at least 10 times less, such as at least 100 times less, and preferably at least 1000 times less, such as 10.000 times less or even less than that, than the K D with which said amino acid sequence or polypeptide binds to the second target or polypeptide.
  • an amino acid sequence or polypeptide when it is "specific for" a first target or antigen compared to a second target or antigen, it is directed against (as defined herein) said first target or antigen, but not directed against said second target or antigen.
  • cross-block when an amino acid sequence or polypeptide is "specific for" a first target or antigen compared to a second target or antigen, it is directed against (as defined herein) said first target or antigen, but not directed against said second target or antigen.
  • cross-block cross-blocked” and “cross-blocking” are used interchangeably herein to mean the ability of an amino acid sequence or other binding agents (such as a polypeptide of the invention) to interfere with the binding of other amino acid sequences or binding agents of the invention to a given target.
  • the extend to which an amino acid sequence or other binding agents of the invention is able to interfere with the binding of another to [target], and therefore whether it can be said to cross-block according to the invention, can be determined using competition binding assays.
  • One particularly suitable quantitative assay uses a Biacore machine which can measure the extent of interactions using surface plasmon resonance technology.
  • Another suitable quantitative cross-blocking assay uses an ELISA-based approach to measure competition between amino acid sequence or another binding agents in terms of their binding to the target.
  • the Biacore machine for example the Biacore 3000
  • the target protein is coupled to a CM5 Biacore chip using standard amine coupling chemistry to generate a surface that is coated with the target.
  • 200- 800 resonance units of the target would be coupled to the chip (an amount that gives easily measurable levels of binding but that is readily saturable by the concentrations of test reagent being used).
  • test amino acid sequences (termed A* and B*) to be assessed for their ability to cross- block each other are mixed at a one to one molar ratio of binding sites in a suitable buffer to create the test mixture.
  • concentrations on a binding site basis the molecular weight of an amino acid sequence is assumed to be the total molecular weight of the amino acid sequence divided by the number of target binding sites on that amino acid sequence.
  • concentration of each amino acid sequence in the test mix should be high enough to readily saturate the binding sites for that amino acid sequence on the target molecules captured on the Biacore chip.
  • the amino acid sequences in the mixture are at the same molar concentration (on a binding basis) and that concentration would typically be between 1.00 and 1.5 micromolar (on a binding site basis).
  • A* and B* in these solutions should be in the same buffer and at the same concentration as in the test mix.
  • the test mixture is passed over the target-coated Biacore chip and the total amount of binding recorded.
  • the chip is then treated in such a way as to remove the bound amino acid sequences without damaging the chip-bound target. Typically this is done by treating the chip with 30 mM HCl for 60 seconds.
  • the solution of A* alone is then passed over the target-coated surface and the amount of binding recorded.
  • the chip is again treated to remove all of the bound amino acid sequences without damaging the chip-bound target.
  • the solution of B* alone is then passed over the target-coated surface and the amount of binding recorded.
  • a cross -blocking amino acid sequence or other binding agent according to the invention is one which will bind to the target in the above Biacore cross-blocking assay such that during the assay and in the presence of a second amino acid sequence or other binding agent of the invention the recorded binding is between 80% and 0.1% (e.g. 80% to 4%) of the maximum theoretical binding, specifically between 75% and 0.1% (e.g.
  • the Biacore assay described above is a primary assay used to determine if amino acid sequences or other binding agents cross-block each other according to the invention. On rare occasions particular amino acid sequences or other binding agents may not bind to target coupled via amine chemistry to a CM5 Biacore chip (this usually occurs when the relevant binding site on target is masked or destroyed by the coupling to the chip).
  • cross-blocking can be determined using a tagged version of the target, for example a N-terminal His-tagged version (R & D Systems, Minneapolis, MN, USA; 2005 cat# 1406-ST-025).
  • a tagged version of the target for example a N-terminal His-tagged version (R & D Systems, Minneapolis, MN, USA; 2005 cat# 1406-ST-025).
  • an anti-His amino acid sequence would be coupled to the Biacore chip and then the His-tagged target would be passed over the surface of the chip and captured by the anti-
  • His amino acid sequence The cross blocking analysis would be carried out essentially as described above, except that after each chip regeneration cycle, new His-tagged target would be loaded back onto the anti-His amino acid sequence coated surface.
  • C-terminal His-tagged target could alternatively be used.
  • various other tags and tag binding protein combinations that are known in the art could be used for such a cross- blocking analysis (e.g. HA tag with anti-HA antibodies; FLAG tag with anti-FLAG antibodies; biotin tag with streptavidin).
  • the general principal of the assay is to have an amino acid sequence or binding agent that is directed against the target coated onto the wells of an ELISA plate. An excess amount of a second, potentially cross-blocking, anti- target amino acid sequence is added in solution (i.e. not bound to the ELISA plate). A limited amount of the target is then added to the wells. The coated amino acid sequence and the amino acid sequence in solution compete for binding of the limited number of target molecules.
  • the plate is washed to remove excess target that has not been bound by the coated amino acid sequence and to also remove the second, solution phase amino acid sequence as well as any complexes formed between the second, solution phase amino acid sequence and target.
  • the amount of bound target is then measured using a reagent that is appropriate to detect the target.
  • An amino acid sequence in solution that is able to cross-block the coated amino acid sequence will be able to cause a decrease in the number of target molecules that the coated amino acid sequence can bind relative to the number of target molecules that the coated amino acid sequence can bind in the absence of the second, solution phase, amino acid sequence.
  • the first amino acid sequence e.g.
  • an Ab-X is chosen to be the immobilized amino acid sequence, it is coated onto the wells of the ELISA plate, after which the plates are blocked with a suitable blocking solution to minimize non-specific binding of reagents that are subsequently added.
  • An excess amount of the second amino acid sequence, i.e. Ab-Y is then added to the ELISA plate such that the moles of Ab-Y [target] binding sites per well are at least 10 fold higher than the moles of Ab-X [target] binding sites that were used, per well, during the coating of the ELISA plate, [target] is then added such that the moles of [target] added per well are at least 25 -fold lower than the moles of Ab-X [target] binding sites that were used for coating each well.
  • the background signal for the assay is defined as the signal obtained in wells with the coated amino acid sequence (in this case Ab-X), second solution phase amino acid sequence (in this case Ab-Y), [target] buffer only (i.e. no target) and target detection reagents.
  • the positive control signal for the assay is defined as the signal obtained in wells with the coated amino acid sequence (in this case Ab-X), second solution phase amino acid sequence buffer only (i.e. no second solution phase amino acid sequence), target and target detection reagents.
  • the ELISA assay may be run in such a manner so as to have the positive control signal be at least 6 times the background signal.
  • the cross-blocking assay may to be run in two formats: 1) format 1 is where Ab-X is the amino acid sequence that is coated onto the ELISA plate and Ab-Y is the competitor amino acid sequence that is in solution and 2) format 2 is where Ab-Y is the amino acid sequence that is coated onto the ELISA plate and Ab-X is the competitor amino acid sequence that is in solution.
  • Ab-X and Ab- Y are defined as cross-blocking if, either in format 1 or in format 2, the solution phase anti-target amino acid sequence is able to cause a reduction of between 60% and 100%, specifically between 70% and 100%, and more specifically between 80% and 100%, of the target detection signal ⁇ i.e. the amount of target bound by the coated amino acid sequence) as compared to the target detection signal obtained in the absence of the solution phase anti- target amino acid sequence (i.e. the positive control wells).
  • the total number of amino acid residues in a Nanobody can be in the region of 110-120, is preferably 112-115, and is most preferably 113.
  • parts, fragments, analogs or derivatives (as further described herein) of a Nanobody are not particularly limited as to their length and/or size, as long as such parts, fragments, analogs or derivatives meet the further requirements outlined herein and are also preferably suitable for the purposes described herein; u)
  • the amino acid residues of a Nanobody are numbered according to the general numbering for V H domains given by Kabat et al. ("Sequence of proteins of immunological interest", US Public Health Services, NIH Bethesda, MD, Publication No. 91), as applied to VHH domains from Camelids in the article of Riechmann and Muyldermans, J. Immunol.
  • FRl of aNanobody comprises the amino acid residues at positions 1-30
  • CDRl of a Nanobody comprises the amino acid residues at positions 31-35
  • FR2 of a Nanobody comprises the amino acids at positions 36-49
  • CDR2 of a Nanobody comprises the amino acid residues at positions 50-65
  • FR3 of aNanobody comprises the amino acid residues at positions 66-94
  • CDR3 of aNanobody comprises the amino acid residues at positions 95-102
  • FR4 of a Nanobody comprises the amino acid residues at positions 103-113.
  • the total number of amino acid residues in each of the CDR' s may vary and may not correspond to the total number of amino acid residues indicated by the Kabat numbering (that is, one or more positions according to the Kabat numbering may not be occupied in the actual sequence, or the actual sequence may contain more amino acid residues than the number allowed for by the Kabat numbering
  • Kabat numbering This means that, generally, the numbering according to Kabat may or may not correspond to the actual numbering of the amino acid residues in the actual sequence. Generally, however, it can be said that, according to the numbering of Kabat and irrespective of the number of amino acid residues in the CDR's, position 1 according to the Kabat numbering corresponds to the start of FRl and vice versa, position 36 according to the Kabat numbering corresponds to the start of FR2 and vice versa, position 66 according to the Kabat numbering corresponds to the start of FR3 and vice versa, and position 103 according to the Kabat numbering corresponds to the start of FR4 and vice versa.].
  • variable domains present in naturally occurring heavy chain antibodies will also be referred to as "V HH domains", in order to distinguish them from the heavy chain variable domains that are present in conventional 4-chain antibodies (which will be referred to hereinbelow as “V H domains”) and from the light chain variable domains that are present in conventional 4-chain antibodies (which will be referred to hereinbelow as "VL domains").
  • VHH domains have a number of unique structural characteristics and functional properties which make isolated V HH domains (as well as Nanobodies based thereon, which share these structural characteristics and functional properties with the naturally occurring VHH domains) and proteins containing the same highly advantageous for use as functional antigen-binding domains or proteins.
  • V HH domains which have been "designed" by nature to functionally bind to an antigen without the presence of, and without any interaction with, a light chain variable domain
  • Nanobodies can function as a single, relatively small, functional antigen-binding structural unit, domain or protein.
  • V HH domains from the VH and VL domains of conventional 4-chain antibodies, which by themselves are generally not suited for practical application as single antigen-binding proteins or domains, but need to be combined in some form or another to provide a functional antigen-binding unit (as in for example conventional antibody fragments such as Fab fragments; in ScFv's fragments, which consist of a VH domain covalently linked to a VL domain).
  • a functional antigen-binding unit as in for example conventional antibody fragments such as Fab fragments; in ScFv's fragments, which consist of a VH domain covalently linked to a VL domain.
  • V HH domains and Nanobodies as single antigen-binding proteins or as antigen-binding domains (i.e. as part of a larger protein or polypeptide) offers a number of significant advantages over the use of conventional V H and V L domains, scFv's or conventional antibody fragments (such as Fab- or F(ab') 2 -fragments): only a single domain is required to bind an antigen with high affinity and with high selectivity, so that there is no need to have two separate domains present, nor to assure that these two domains are present in the right spatial conformation and configuration (i.e. through the use of especially designed linkers, as with scFv's); - VHH domains and Nanobodies can be expressed from a single gene and require no post- translational folding or modifications;
  • V HH domains and Nanobodies can easily be engineered into multivalent and multispecific formats (as further discussed herein);
  • V HH domains and Nanobodies are highly soluble and do not have a tendency to aggregate (as with the mouse-derived "dAb's" described by Ward et al., Nature, Vol.
  • VHH domains and Nanobodies are highly stable to heat, pH, proteases and other denaturing agents or conditions (see for example Ewert et al, supra); VHH domains and Nanobodies are easy and relatively cheap to prepare, even on a scale required for production.
  • VHH domains, Nanobodies and proteins/polypeptides containing the same can be produced using microbial fermentation (e.g.
  • V HH domains and Nanobodies are relatively small (approximately 15 kDa, or 10 times smaller than a conventional IgG) compared to conventional 4-chain antibodies and antigen-binding fragments thereof, and therefore show high(er) penetration into tissues (including but not limited to solid tumors and other dense tissues) than such conventional 4-chain antibodies and antigen-binding fragments thereof; VHH domains and Nanobodies can show so-called cavity-binding properties (inter alia due to their extended CDR3 loop, compared to conventional V H domains) and can therefore also access targets and epitopes not accessible to conventional 4-chain antibodies and antigen-binding fragments thereof. For example, it has been shown that VHH domains and Nanobodies can inhibit enzymes (see for example WO 97/49805;
  • the invention provides Nanobodies against a metalloproteinase from the ADAM family, and in particular Nanobodies against a metalloproteinase from the ADAM family from a warm-blooded animal, and more in particular Nanobodies against a metalloproteinase from the ADAM family from a mammal, and especially Nanobodies against human a metalloproteinase from the ADAM family; as well as proteins and/or polypeptides comprising at least one such Nanobody.
  • the invention provides Nanobodies against a metalloproteinase from the ADAM family, and proteins and/or polypeptides comprising the same, that have improved therapeutic and/or pharmacological properties and/or other advantageous properties (such as, for example, improved ease of preparation and/or reduced costs of goods), compared to conventional antibodies against a metalloproteinase from the ADAM family or fragments thereof, compared to constructs that could be based on such conventional antibodies or antibody fragments (such as Fab' fragments, F(ab') 2 fragments, ScFv constructs, "diabodies” and other multispecific constructs (see for example the review by Holliger and Hudson, Nat Biotechnol.
  • the Nanobodies of the invention are preferably in essentially isolated form (as defined herein), or form part of a protein or polypeptide of the invention (as defined herein), which may comprise or essentially consist of one or more Nanobodies of the invention and which may optionally further comprise one or more further amino acid sequences (all optionally linked via one or more suitable linkers).
  • the one or more amino acid sequences of the invention may be used as a binding unit in such a protein or polypeptide, which may optionally contain one or more further amino acid sequences that can serve as a binding unit (i.e.
  • such a protein or polypeptide may comprise or essentially consist of one or more Nanobodies of the invention and optionally one or more (other) Nanobodies (i.e. directed against other targets than a metalloproteinase from the ADAM family), all optionally linked via one or more suitable linkers, so as to provide a monovalent, multivalent or multispecific Nanobody construct, respectively, as further described herein.
  • Such proteins or polypeptides may also be in essentially isolated form (as defined herein).
  • the binding site for binding against a metalloproteinase from the ADAM family is preferably formed by the CDR sequences.
  • a Nanobody of the invention may also, and in addition to the at least one binding site for binding against a metalloproteinase from the ADAM family, contain one or more further binding sites for binding against other antigens, proteins or targets.
  • a Nanobody of the invention when a Nanobody of the invention (or a polypeptide of the invention comprising the same) is intended for administration to a subject (for example for therapeutic and/or diagnostic purposes as described herein), it is preferably directed against human a metalloproteinase from the ADAM family; whereas for veterinary purposes, it is preferably directed against a metalloproteinase from the ADAM family from the species to be treated.
  • a Nanobody of the invention may or may not be cross-reactive (i.e.
  • a metalloproteinase from the ADAM family from two or more species of mammal, such as against human a metalloproteinase from the ADAM family and a metalloproteinase from the ADAM family from at least one of the species of mammal mentioned herein).
  • the Nanobodies of the invention may generally be directed against any antigenic determinant, epitope, part, domain, subunit or confirmation (where applicable) of a metalloproteinase from the ADAM family.
  • the Nanobodies of the invention and polypeptides comprising the same) are directed against one of the catalytic sites or binding sites mentioned generally herein for the amino acid sequences of the invention.
  • the amino acid sequence and structure of a Nanobody can be considered - without however being limited thereto - to be comprised of four framework regions or "FR' s" (or sometimes also referred to as “FWs"), which are referred to in the art and herein as “Framework region 1" or “FRl”; as “Framework region 2" or “FR2”; as “Framework region 3" or “FR3”; and as “Framework region 4" or “FR4", respectively; which framework regions are interrupted by three complementary determining regions or "CDR's", which are referred to in the art as “Complementarity Determining Region l”or “CDRl”; as “Complementarity Determining Region 2" or “CDR2”; and as “Complementarity Determining Region 3" or “CDR3", respectively.
  • CDR's complementary determining regions
  • the Nanobodies of the invention are such that: the Nanobodies can bind to a metalloproteinase from the ADAM family with a dissociation constant (K D ) of 10 "5 to IO 42 moles/liter or less, and preferably 10 "7 to 10 "12 moles/liter or less and more preferably 10 " to 10 " moles/liter (i.e.
  • K D dissociation constant
  • the Nanobodies can bind to a metalloproteinase from the ADAM family with a k on -rate of between 10 2 M 4 S "1 to about 10 7 M ' V 1 , preferably between 10 3 M ' V 1 and 10 7 M 4 S "1 , more preferably between 10 4 M 4 S "1 and 10 7 M 4 S "1 , such as between 10 5 M -1 S 4 and 10 7
  • the CDR sequences present in) the Nanobodies of the invention are such that: a monovalent Nanobody of the invention (or a polypeptide that contains only one Nanobody of the invention) is preferably such that it will bind to a metalloproteinase from the ADAM family with an affinity less than 500 nM, preferably less than 200 nM, more preferably less than 10 nM, such as less than 500 pM.
  • the affinity of the Nanobody of the invention against a metalloproteinase from the ADAM family can be determined in a manner known per se, for example using the general techniques for measuring KD. KA, k o ff or k on mentioned herein, as well as some of the specific assays described herein.
  • Nanobodies of the invention and of polypeptides comprising the same
  • a metalloproteinase from the ADAM family will become clear from the further description and examples herein.
  • the invention relates to a Nanobody (as defined herein) against a metalloproteinase from the ADAM family, which consists of 4 framework regions (FRl to FR4 respectively) and 3 complementarity determining regions (CDRl to CDR3 respectively), in which: - CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 337; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 232 to 337; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 232 to 337; and/or
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 444 to 549; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 444 to 549; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 444 to 549; and/or
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 656 to 761; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 656 to 761; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 656 to 761; or any suitable fragment of such an amino acid sequence.
  • the invention relates to a Nanobody (as defined herein) against a metalloproteinase from the ADAM family, which consists of 4 framework regions (FRl to FR4 respectively) and 3 complementarity determining regions (CDRl to CDR3 respectively), in which:
  • CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 337; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 232 to 337; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 232 to 337; and - CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 444 to 549; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 444 to 549; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 444 to 549; and
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 656 to 761; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 656 to 761; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 656 to 761; or any suitable fragment of such an amino acid sequences.
  • the invention relates to a Nanobody (as defined herein) against a ADAM8, which consists of 4 framework regions (FRl to FR4 respectively) and 3 complementarity determining regions (CDRl to CDR3 respectively), in which: CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 250; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 232 to 250; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 232 to 250; and/or CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 444 to 462; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 444 to 4
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 656 to 674; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 656 to 674; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 656 to 674; or any suitable fragment of such an amino acid sequence.
  • the invention relates to a Nanobody (as defined herein) ADAM8, which consists of 4 framework regions (FRl to FR4 respectively) and 3 complementarity determining regions (CDRl to CDR3 respectively), in which:
  • CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 232 to 250; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 232 to 250; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 232 to 250; and
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 444 to 462; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 444 to 462; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 444 to 462; and
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 656 to 674; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 656 to 674; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 656 to 674; or any suitable fragment of such an amino acid sequences.
  • the invention relates to a Nanobody (as defined herein) against a ADAM9, which consists of 4 framework regions (FRl to FR4 respectively) and 3 complementarity determining regions (CDRl to CDR3 respectively), in which: - CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; and/or
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; and/or
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027; or any suitable fragment of such an amino acid sequence.
  • the invention relates to a Nanobody (as defined herein) ADAM9, which consists of 4 framework regions (FRl to FR4 respectively) and 3 complementarity determining regions (CDRl to CDR3 respectively), in which:
  • CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 251 to 271 and/or 984 to 987; and - CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 463 to 483 and/or 1004 to 1007; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 675 to 695 and/or 1024 to 1027; or any suitable fragment of such an amino acid sequences.
  • the invention relates to a Nanobody (as defined herein) against a ADAMlO, which consists of 4 framework regions (FRl to FR4 respectively) and 3 complementarity determining regions (CDRl to CDR3 respectively), in which:
  • CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 272 to 295; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 272 to 295; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 272 to 295; and/or
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 484 to 507; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 484 to 507; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 484 to 507; and/or
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 696 to 719; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 696 to 719; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 696 to 719; or any suitable fragment of such an amino acid sequence.
  • the invention relates to a Nanobody (as defined herein) ADAMlO, which consists of 4 framework regions (FRl to FR4 respectively) and 3 complementarity determining regions (CDRl to CDR3 respectively), in which: - CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 272 to 295; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 272 to 295; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 272 to 295 ; and
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 484 to 507; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 484 to 507; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 484 to 507; and CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 696 to 719; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 696 to 719; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 696 to 719; or any suitable fragment of such an amino acid sequences.
  • the invention relates to a Nanobody (as defined herein) against a ADAM 17, which consists of 4 framework regions (FRl to FR4 respectively) and 3 complementarity determining regions (CDRl to CDR3 respectively), in which:
  • CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; and/or
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; and/or
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032; or any suitable fragment of such an amino acid sequence.
  • the invention relates to a Nanobody (as defined herein) ADAM 17, which consists of 4 framework regions (FRl to FR4 respectively) and 3 complementarity determining regions (CDRl to CDR3 respectively), in which: - CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 296 to 312 and/or 988 to 992; and
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 508 to 524 and/or 1008 to 1012; and
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 720 to 736 and/or 1028 to 1032; or any suitable fragment of such an amino acid sequences.
  • the invention relates to a Nanobody (as defined herein) against a ADAMTS5, which consists of 4 framework regions (FRl to FR4 respectively) and 3 complementarity determining regions (CDRl to CDR3 respectively), in which: - CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 313 to 337 and/or 993; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 313 to 337 and/or 993; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 313 to 337 and/or 993; and/or
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 525 to 549 and/or 1013; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 525 to 549 and/or 1013; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 525 to 549 and/or 1013; and/or
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 737 to 761 and/or 1033; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 737 to 761 and/or 1033; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 737 to 761 and/or 1033; or any suitable fragment of such an amino acid sequence.
  • the invention relates to a Nanobody (as defined herein) ADAMTS5, which consists of 4 framework regions (FRl to FR4 respectively) and 3 complementarity determining regions (CDRl to CDR3 respectively), in which:
  • CDRl is chosen from the group consisting of: a) the amino acid sequences of SEQ ID NO's: 313 to 337 and/or 993; b) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 313 to 337 and/or 993; c) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 313 to 337 and/or 993; and
  • CDR2 is chosen from the group consisting of: d) the amino acid sequences of SEQ ID NO's: 525 to 549 and/or 1013; e) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 525 to 549 and/or 1013; f) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 525 to 549 and/or 1013; and
  • CDR3 is chosen from the group consisting of: g) the amino acid sequences of SEQ ID NO's: 737 to 761 and/or 1033; h) amino acid sequences that have at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 737 to 761 and/or 1033; i) amino acid sequences that have 3, 2, or 1 amino acid difference with at least one of the amino acid sequences of SEQ ID NO's: 737 to 761 and/or 1033; or any suitable fragment of such an amino acid sequences.
  • any amino acid substitution in such a CDR according to b) and/or c) is preferably, and compared to the corresponding CDR according to a), a conservative amino acid substitution (as defined herein); and/or ii) the CDR according to b) and/or c) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding CDR according to a); and/or iii) the CDR according to b) and/or c) may be a CDR that is derived from a CDR according to a) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • any amino acid substitution in such a CDR according to e) and/or f) is preferably, and compared to the corresponding CDR according to d), a conservative amino acid substitution (as defined herein); and/or ii) the CDR according to e) and/or f) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding CDR according to d); and/or iii) the CDR according to e) and/or f) may be a CDR that is derived from a CDR according to d) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • any amino acid substitution in such a CDR according to h) and/or i) is preferably, and compared to the corresponding CDR according to g), a conservative amino acid substitution (as defined herein); and/or ii) the CDR according to h) and/or i) preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding CDR according to g); and/or iii) the CDR according to h) and/or i) may be a CDR that is derived from a CDR according to g) by means of affinity maturation using one or more techniques of affinity maturation known per se.
  • Nanobody of the invention that comprises one or more CDRl sequences, CDR2 sequences and/or CDR3 sequences according to b), c), e), f), h) or i), respectively.
  • Nanobodies comprising one or more of the CDR' s explicitly listed above are particularly preferred; Nanobodies comprising two or more of the CDR's explicitly listed above are more particularly preferred; and Nanobodies comprising three of the CDR's explicitly listed above are most particularly preferred.
  • Some particularly preferred, but non-limiting combinations of CDR sequences, as well as preferred combinations of CDR sequences and framework sequences, are mentioned in Table A-I below, which lists the CDR sequences and framework sequences that are present in a number of preferred (but non-limiting) Nanobodies of the invention.
  • Table A-I lists the CDR sequences and framework sequences that are present in a number of preferred (but non-limiting) Nanobodies of the invention.
  • a combination of CDRl, CDR2 and CDR3 sequences that occur in the same clone i.e.
  • CDRl, CDR2 and CDR3 sequences that are mentioned on the same line in Table A-I will usually be preferred (although the invention in its broadest sense is not limited thereto, and also comprises other suitable combinations of the CDR sequences mentioned in Table A-I).
  • a combination of CDR sequences and framework sequences that occur in the same clone i.e. CDR sequences and framework sequences that are mentioned on the same line in Table A-I
  • will usually be preferred although the invention in its broadest sense is not limited thereto, and also comprises other suitable combinations of the CDR sequences and framework sequences mentioned in Table A-I, as well as combinations of such CDR sequences and other suitable framework sequences, e.g. as further described herein).
  • each CDR can be replaced by a CDR chosen from the group consisting of amino acid sequences that have at least 80%, preferably at least 90%, more preferably at least 95%, even more preferably at least 99% sequence identity (as defined herein) with the mentioned CDR's; in which: i) any amino acid substitution in such a CDR is preferably, and compared to the corresponding CDR sequence mentioned in Table A-I, a conservative amino acid substitution (as defined herein); and/or ii) any such CDR sequence preferably only contains amino acid substitutions, and no amino acid deletions or insertions, compared to the corresponding CDR sequence mentioned in Table A-I; and/or iii) any such CDR sequence is a CDR that is derived by means of a technique for affinity maturation known per se, and in particular starting from the corresponding CDR sequence mentioned in Table A-I.
  • At least one of the CDRl, CDR2 and CDR3 sequences present is suitably chosen from the group consisting of the CDRl, CDR2 and CDR3 sequences, respectively, listed in Table A-I; or from the group of CDRl, CDR2 and CDR3 sequences, respectively, that have at least 80%, preferably at least 90%, more preferably at least 95%, even more preferably at least 99% "sequence identity" (as defined herein) with at least one of the CDRl, CDR2 and CDR3 sequences, respectively, listed in Table A-I; and/or from the group consisting of the CDRl, CDR2 and CDR3 sequences, respectively, that have 3, 2 or only 1 "amino acid difference(s)" (as defined herein) with at least one of the CDRl, CDR2 and CDR3 sequences, respectively, listed in Table A-I.
  • suitably chosen is meant that, as applicable, a CDRl sequence is chosen from suitable CDRl sequences (i.e. as defined herein), a CDR2 sequence is chosen from suitable CDR2 sequences (i.e. as defined herein), and a CDR3 sequence is chosen from suitable CDR3 sequence (i.e. as defined herein), respectively.
  • the CDR sequences are preferably chosen such that the Nanobodies of the invention bind to a metalloproteinase from the ADAM family with an affinity (suitably measured and/or expressed as a K D -value (actual or apparent), a K A -value (actual or apparent), a k on -rate and/or a k o ff-rate, or alternatively as an IC50 value, as further described herein) that is as defined herein.
  • At least the CDR3 sequence present is suitably chosen from the group consisting of the CDR3 sequences listed in Table A-I or from the group of CDR3 sequences that have at least 80%, preferably at least 90%, more preferably at least 95%, even more preferably at least 99% sequence identity with at least one of the CDR3 sequences listed in Table A-I; and/or from the group consisting of the CDR3 sequences that have 3, 2 or only 1 amino acid difference(s) with at least one of the CDR3 sequences listed in Table A-I.
  • At least two of the CDRl, CDR2 and CDR3 sequences present are suitably chosen from the group consisting of the CDRl, CDR2 and CDR3 sequences, respectively, listed in Table A-I or from the group consisting of CDRl, CDR2 and CDR3 sequences, respectively, that have at least 80%, preferably at least 90%, more preferably at least 95%, even more preferably at least 99% sequence identity with at least one of the CDRl, CDR2 and CDR3 sequences, respectively, listed in Table A-I; and/or from the group consisting of the CDRl, CDR2 and CDR3 sequences, respectively, that have 3, 2 or only 1 "amino acid difference(s)" with at least one of the CDRl, CDR2 and CDR3 sequences, respectively, listed in Table A-I .
  • At least the CDR3 sequence present is suitably chosen from the group consisting of the CDR3 sequences listed in Table A-I or from the group of CDR3 sequences that have at least 80%, preferably at least 90%, more preferably at least 95%, even more preferably at least 99% sequence identity with at least one of the CDR3 sequences listed in Table A-I, respectively; and at least one of the CDRl and CDR2 sequences present is suitably chosen from the group consisting of the CDRl and CDR2 sequences, respectively, listed in Table A-I or from the group of CDRl and CDR2 sequences, respectively, that have at least 80%, preferably at least 90%, more preferably at least 95%, even more preferably at least 99% sequence identity with at least one of the CDRl and CDR2 sequences, respectively, listed in Table A-I; and/or from the group consisting of the CDRl and CDR2 sequences, respectively, that have 3, 2 or only 1 amino acid difference(s
  • all three CDRl, CDR2 and CDR3 sequences present are suitably chosen from the group consisting of the CDRl, CDR2 and CDR3 sequences, respectively, listed in Table A-I or from the group of CDRl, CDR2 and CDR3 sequences, respectively, that have at least 80%, preferably at least 90%, more preferably at least 95%, even more preferably at least 99% sequence identity with at least one of the CDRl, CDR2 and CDR3 sequences, respectively, listed in Table A-I; and/or from the group consisting of the CDRl, CDR2 and CDR3 sequences, respectively, that have 3, 2 or only 1 amino acid difference(s) with at least one of the CDRl, CDR2 and CDR3 sequences, respectively, listed in Table A-I. Even more preferably, in the Nanobodies of the invention, at least one of the CDRl,
  • CDR2 and CDR3 sequences present is suitably chosen from the group consisting of the CDRl, CDR2 and CDR3 sequences, respectively, listed in Table A-I.
  • at least one or preferably both of the other two CDR sequences present are suitably chosen from CDR sequences that have at least 80%, preferably at least 90%, more preferably at least 95%, even more preferably at least 99% sequence identity with at least one of the corresponding CDR sequences, respectively, listed in Table A-I; and/or from the group consisting of the CDR sequences that have 3, 2 or only 1 amino acid difference(s) with at least one of the corresponding sequences, respectively, listed in Table A-I.
  • the CDR3 sequence present is suitably chosen from the group consisting of the CDR3 listed in Table A-I.
  • at least one and preferably both of the CDRl and CDR2 sequences present are suitably chosen from the groups of CDRl and CDR2 sequences, respectively, that have at least 80%, preferably at least 90%, more preferably at least 95%, even more preferably at least 99% sequence identity with the CDRl and CDR2 sequences, respectively, listed in Table A-I; and/or from the group consisting of the CDRl and CDR2 sequences, respectively, that have 3, 2 or only 1 amino acid difference(s) with at least one of the CDRl and CDR2 sequences, respectively, listed in Table A-I.
  • at least two of the CDRl are suitably chosen from the groups of CDRl and CDR2 sequences, respectively, that have at least 80%, preferably at least 90%, more preferably at least 95%, even more preferably at least 99% sequence identity with the CDRl and CDR2 sequences, respectively
  • CDR2 and CDR3 sequences present are suitably chosen from the group consisting of the CDRl, CDR2 and CDR3 sequences, respectively, listed in Table A-I.
  • the remaining CDR sequence present is suitably chosen from the group of CDR sequences that have at least 80%, preferably at least 90%, more preferably at least 95%, even more preferably at least 99% sequence identity with at least one of the corresponding CDR sequences listed in Table A-I; and/or from the group consisting of CDR sequences that have 3, 2 or only 1 amino acid difference(s) with at least one of the corresponding sequences listed in Table A-I.
  • the CDR3 sequence is suitably chosen from the group consisting of the CDR3 sequences listed in Table A-I, and either the CDRl sequence or the CDR2 sequence is suitably chosen from the group consisting of the CDRl and CDR2 sequences, respectively, listed in Table A-I.
  • the remaining CDR sequence present is suitably chosen from the group of CDR sequences that have at least 80%, preferably at least 90%, more preferably at least 95%, even more preferably at least 99% sequence identity with at least one of the corresponding CDR sequences listed in Table A-I; and/or from the group consisting of CDR sequences that have 3, 2 or only 1 amino acid difference(s) with the corresponding CDR sequences listed in Table A-I.
  • a CDR in aNanobody of the invention is a CDR sequence mentioned in Table A-I or is suitably chosen from the group of CDR sequences that have at least 80%, preferably at least 90%, more preferably at least 95%, even more preferably at least 99% sequence identity with a CDR sequence listed in Table A-I; and/or from the group consisting of CDR sequences that have 3, 2 or only 1 amino acid difference(s) with a CDR sequence listed in Table A-I, that at least one and preferably both of the other CDR' s are suitably chosen from the CDR sequences that belong to the same combination in Table A-I (i.e.
  • a Nanobody of the invention can for example comprise a CDRl sequence that has more than 80 % sequence identity with one of the CDRl sequences mentioned in Table A-I, a CDR2 sequence that has 3, 2 or 1 amino acid difference with one of the CDR2 sequences mentioned in Table A-I (but belonging to a different combination), and a CDR3 sequence.
  • Nanobodies of the invention may for example comprise: (1) a CDRl sequence that has more than 80 % sequence identity with one of the CDRl sequences mentioned in Table A-I; a CDR2 sequence that has 3, 2 or 1 amino acid difference with one of the CDR2 sequences mentioned in Table A-I (but belonging to a different combination); and a CDR3 sequence that has more than 80 % sequence identity with one of the CDR3 sequences mentioned in Table A-I (but belonging to a different combination); or (2) a CDRl sequence that has more than 80 % sequence identity with one of the CDRl sequences mentioned in Table A-I; a CDR2 sequence, and one of the CDR3 sequences listed in Table A-I; or (3) a CDRl sequence; a CDR2 sequence that has more than 80% sequence identity with one of the CDR2 sequence listed in Table A-I; and a CDR3 sequence that has 3, 2 or 1 amino acid differences with the CDR3 sequence mentioned in Table A-I that belongs to the same c ;
  • Nanobodies of the invention may for example comprise: ⁇ DRl sequence that has more than 80 % sequence identity with one of the CDRl sequences mentioned in Table A-I; a CDR2 sequence that has 3, 2 or 1 amino acid difference with the CDR2 sequence mentioned in Table A-I that belongs to the same combination; and a CDR3 sequence that has more than 80 % sequence identity with the CDR3 sequence mentioned in Table A-I that belongs to the same combination; (2) a CDRl sequence; a CDR 2 listed in Table A-I and a CDR3 sequence listed in Table A-I (in which the CDR2 sequence and CDR3 sequence may belong to different combinations).
  • Nanobodies of the invention may for example comprise: (1) a CDRl sequence that has more than 80 % sequence identity with one of the CDRl sequences mentioned in Table A-I; the CDR2 sequence listed in Table A-I that belongs to the same combination; and a CDR3 sequence mentioned in Table A-I that belongs to a different combination; or (2) a CDRl sequence mentioned in Table A-I; a CDR2 sequence that has 3, 2 or 1 amino acid differences with the CDR2 sequence mentioned in Table A-I that belongs to the same combination; and a CDR3 sequence that has more than 80% sequence identity with the CDR3 sequence listed in Table A-I that belongs to the same or a different combination.
  • Nanobodies of the invention may for example comprise a CDRl sequence mentioned in Table A-I, a CDR2 sequence that has more than 80 % sequence identity with the CDR2 sequence mentioned in Table A-I that belongs to the same combination; and the CDR3 sequence mentioned in Table A-I that belongs to the same combination.
  • the CDRl, CDR2 and CDR3 sequences present are suitably chosen from one of the combinations of CDRl, CDR2 and CDR3 sequences, respectively, listed in Table A-I .
  • CDRl has a length of between 1 and 12 amino acid residues, and usually between 2 and 9 amino acid residues, such as 5, 6 or 7 amino acid residues; and/or (b) CDR2 has a length of between 13 and 24 amino acid residues, and usually between 15 and 21 amino acid residues, such as 16 and 17 amino acid residues; and/or (c) CDR3 has a length of between 2 and 35 amino acid residues, and usually between 3 and 30 amino acid residues, such as between 6 and 23 amino acid residues.
  • the invention relates to a Nanobody in which the CDR sequences (as defined herein) have more than 80%, preferably more than 90%, more preferably more than 95%, such as 99% or more sequence identity (as defined herein) with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 868 to 973 and/or 1044 to 1053.
  • Nanobodies with the above CDR sequences may be as further described herein, and preferably have framework sequences that are also as further described herein.
  • such Nanobodies may be naturally occurring Nanobodies (from any suitable species), naturally occurring V HH sequences (i.e. from a suitable species of Camelid) or synthetic or semi-synthetic amino acid sequences or Nanobodies, including but not limited to partially humanized Nanobodies or VHH sequences, fully humanized Nanobodies or V HH sequences, camelized heavy chain variable domain sequences, as well as Nanobodies that have been obtained by the techniques mentioned herein.
  • the invention relates to a humanized Nanobody, which consists of 4 framework regions (FRl to FR4 respectively) and 3 complementarity determining regions (CDRl to CDR3 respectively), in which CDRl to CDR3 are as defined herein and in which said humanized Nanobody comprises at least one humanizing substitution (as defined herein), and in particular at least one humanizing substitution in at least one of its framework sequences (as defined herein).
  • the invention relates to a Nanobody in which the CDR sequences have at least 70% amino acid identity, preferably at least 80% amino acid identity, more preferably at least 90% amino acid identity, such as 95% amino acid identity or more or even essentially 100% amino acid identity with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 868 to 973 and/or 1044 to 1053.
  • This degree of amino acid identity can for example be determined by determining the degree of amino acid identity (in a manner described herein) between said Nanobody and one or more of the sequences of SEQ ID NO's: 868 to 973 and/or 1044 to 1053, in which the amino acid residues that form the framework regions are disregarded.
  • Such Nanobodies can be as further described herein.
  • the invention relates to a Nanobody with an amino acid sequence that is chosen from the group consisting of SEQ ID NO's: 868 to 973 and/or 1044 to 1053 or from the group consisting of from amino acid sequences that have more than 80%, preferably more than 90%, more preferably more than 95%, such as 99% or more sequence identity (as defined herein) with at least one of the amino acid sequences of SEQ ID NO's: 868 to 973 and/or 1044 to 1053.
  • Another preferred, but non-limiting aspect of the invention relates to humanized variants of the Nanobodies of SEQ ID NO's: 868 to 973 and/or 1044 to 1053, that comprise, compared to the corresponding native V HH sequence, at least one humanizing substitution (as defined herein), and in particular at least one humanizing substitution in at least one of its framework sequences (as defined herein).
  • polypeptides of the invention comprise or essentially consist of at least one Nanobody of the invention.
  • Nanobodies that are mentioned herein as “preferred” (or “more preferred”, “even more preferred”, etc.) are also preferred (or more preferred, or even more preferred, etc.) for use in the polypeptides described herein.
  • polypeptides that comprise or essentially consist of one or more "preferred” Nanobodies of the invention will generally be preferred, and polypeptides that comprise or essentially consist of one or more "more preferred” Nanobodies of the invention will generally be more preferred, etc..
  • Nanobody (such as a single Nanobody of the invention) will be referred to herein as "monovalent” proteins or polypeptides or as “monovalent constructs”. Proteins and polypeptides that comprise or essentially consist of two or more Nanobodies (such as at least two Nanobodies of the invention or at least one Nanobody of the invention and at least one other Nanobody) will be referred to herein as “multivalent” proteins or polypeptides or as “multivalent constructs”, and these may provide certain advantages compared to the corresponding monovalent Nanobodies of the invention. Some non-limiting examples of such multivalent constructs will become clear from the further description herein.
  • a polypeptide of the invention comprises or essentially consists of at least two Nanobodies of the invention, such as two or three Nanobodies of the invention.
  • multivalent constructs can provide certain advantages compared to a protein or polypeptide comprising or essentially consisting of a single Nanobody of the invention, such as a much improved avidity for a metalloproteinase from the ADAM family.
  • Such multivalent constructs will be clear to the skilled person based on the disclosure herein.
  • a polypeptide of the invention comprises or essentially consists of at least one Nanobody of the invention and at least one other binding unit (i.e. directed against another epitope, antigen, target, protein or polypeptide), which is preferably also a Nanobody.
  • Such proteins or polypeptides are also referred to herein as "multispecific” proteins or polypeptides or as 'multispecific constructs", and these may provide certain advantages compared to the corresponding monovalent Nanobodies of the invention (as will become clear from the further discussion herein of some preferred, but-nonlimiting multispecific constructs). Such multispecific constructs will be clear to the skilled person based on the disclosure herein.
  • a polypeptide of the invention comprises or essentially consists of at least one Nanobody of the invention, optionally one or more further Nanobodies, and at least one other amino acid sequence (such as a protein or polypeptide) that confers at least one desired property to the Nanobody of the invention and/or to the resulting fusion protein.
  • at least one other amino acid sequence such as a protein or polypeptide
  • such fusion proteins may provide certain advantages compared to the corresponding monovalent Nanobodies of the invention.
  • the one or more Nanobodies and/or other amino acid sequences may be directly linked to each other and/or suitably linked to each other via one or more linker sequences.
  • linker sequences Some suitable but non-limiting examples of such linkers will become clear from the further description herein.
  • a Nanobody of the invention or a compound, construct or polypeptide of the invention comprising at least one Nanobody of the invention may have an increased half-life, compared to the corresponding amino acid sequence of the invention.
  • Nanobodies, compounds and polypeptides will become clear to the skilled person based on the further disclosure herein, and for example comprise Nanobodies sequences or polypeptides of the invention that have been chemically modified to increase the half-life thereof (for example, by means of pegylation); amino acid sequences of the invention that comprise at least one additional binding site for binding to a serum protein (such as serum albumin, see for example EP 0 368 684 Bl, page 4); or polypeptides of the invention that comprise at least one Nanobody of the invention that is linked to at least one moiety (and in particular at least one amino acid sequence) that increases the half-life of the Nanobody of the invention.
  • a serum protein such as serum albumin, see for example EP 0 368 684 Bl, page 4
  • polypeptides of the invention that comprise at least one Nanobody of the invention that is linked to at least one moiety (and in particular at least one amino acid sequence) that increases the half-life of the Nanobody of the invention.
  • polypeptides of the invention that comprise such half-life extending moieties or amino acid sequences will become clear to the skilled person based on the further disclosure herein; and for example include, without limitation, polypeptides in which the one or more Nanobodies of the invention are suitable linked to one or more serum proteins or fragments thereof (such as serum albumin or suitable fragments thereof) or to one or more binding units that can bind to serum proteins (such as, for example, Nanobodies or (single) domain antibodies that can bind to serum proteins such as serum albumin, serum immunoglobulins such as IgG, or transferrin); polypeptides in which a Nanobody of the invention is linked to an Fc portion (such as a human Fc) or a suitable part or fragment thereof; or polypeptides in which the one or more Nanobodies of the invention are suitable linked to one or more small proteins or peptides that can bind to serum proteins (such as, without limitation, the proteins and peptides described in WO 91/01743, WO 01
  • Nanobodies, compounds, constructs or polypeptides may contain one or more additional groups, residues, moieties or binding units, such as one or more further amino acid sequences and in particular one or more additional Nanobodies (i.e. not directed against a metalloproteinase from the ADAM family), so as to provide a tri- of multispecific Nanobody construct.
  • the Nanobodies of the invention (or compounds, constructs or polypeptides comprising the same) with increased half-life preferably have a half-life that is at least 1.5 times, preferably at least 2 times, such as at least 5 times, for example at least 10 times or more than 20 times, greater than the half-life of the corresponding amino acid sequence of the invention per se.
  • the Nanobodies, compounds, constructs or polypeptides of the invention with increased half-life may have a half-life that is increased with more than 1 hours, preferably more than 2 hours, more preferably more than 6 hours, such as more than 12 hours, or even more than 24, 48 or 72 hours, compared to the corresponding amino acid sequence of the invention per se.
  • Nanobodies, compound, constructs or polypeptides of the invention exhibit a serum half-life in human of at least about 12 hours, preferably at least 24 hours, more preferably at least 48 hours, even more preferably at least 72 hours or more.
  • compounds or polypeptides of the invention may have a half-life of at least 5 days (such as about 5 to 10 days), preferably at least 9 days (such as about 9 to 14 days), more preferably at least about 10 days (such as about 10 to 15 days), or at least about 11 days (such as about 11 to 16 days), more preferably at least about 12 days (such as about 12 to 18 days or more), or more than 14 days (such as about 14 to 19 days).
  • a polypeptide of the invention comprises one or more (such as two or preferably one) Nanobodies of the invention linked (optionally via one or more suitable linker sequences) to one or more (such as two and preferably one) amino acid sequences that allow the resulting polypeptide of the invention to cross the blood brain barrier.
  • said one or more amino acid sequences that allow the resulting polypeptides of the invention to cross the blood brain barrier may be one or more (such as two and preferably one) Nanobodies, such as the Nanobodies described in WO 02/057445, of which FC44 (SEQ ID NO: 189 of WO 06/040153) and FC5 (SEQ ID NO: 190 of WO 06/040154) are preferred examples.
  • polypeptides comprising one or more Nanobodies of the invention are preferably such that they: bind to a metalloproteinase from the ADAM family with a dissociation constant (KD) of 10 "5 to 10 "12 moles/liter or less, and preferably 10 "7 to IO 42 moles/liter or less and more preferably 10 "8 to 10 "12 moles/liter (i.e.
  • KD dissociation constant
  • K A
  • a polypeptide that contains only one amino acid sequence of the invention is preferably such that it will bind to a metalloproteinase from the ADAM family with an affinity less than 500 nM, preferably less than 200 nM, more preferably less than 10 nM, such as less than 500 pM.
  • a polypeptide that contains two or more Nanobodies of the invention may bind to a metalloproteinase from the ADAM family with an increased avidity, compared to a polypeptide that contains only one amino acid sequence of the invention.
  • IC 50 values for binding of the amino acid sequences or polypeptides of the invention to a metalloproteinase from the ADAM family will become clear from the further description and examples herein.
  • nucleic acid that encodes an amino acid sequence of the invention (such as a Nanobody of the invention) or a polypeptide of the invention comprising the same.
  • an amino acid sequence of the invention such as a Nanobody of the invention
  • polypeptide of the invention comprising the same.
  • such a nucleic acid may be in the form of a genetic construct, as defined herein.
  • the invention relates to host or host cell that expresses or that is capable of expressing an amino acid sequence (such as a Nanobody) of the invention and/or a polypeptide of the invention comprising the same; and/or that contains a nucleic acid of the invention.
  • an amino acid sequence such as a Nanobody
  • a polypeptide of the invention comprising the same; and/or that contains a nucleic acid of the invention.
  • Another aspect of the invention relates to a product or composition containing or comprising at least one amino acid sequence of the invention, at least one polypeptide of the invention and/or at least one nucleic acid of the invention, and optionally one or more further components of such compositions known per se, i.e. depending on the intended use of the composition.
  • a product or composition may for example be a pharmaceutical composition (as described herein), a veterinary composition or a product or composition for diagnostic use (as also described herein).
  • the invention further relates to methods for preparing or generating the amino acid sequences, compounds, constructs, polypeptides, nucleic acids, host cells, products and compositions described herein. Some preferred but non-limiting examples of such methods will become clear from the further description herein.
  • the invention further relates to applications and uses of the amino acid sequences, compounds, constructs, polypeptides, nucleic acids, host cells, products and compositions described herein, as well as to methods for the prevention and/or treatment for diseases and disorders associated with a metalloproteinase from the ADAM family. Some preferred but non-limiting applications and uses will become clear from the further description herein.
  • Nanobody as used herein in its broadest sense is not limited to a specific biological source or to a specific method of preparation.
  • the Nanobodies of the invention can generally be obtained: (1) by isolating the VHH domain of a naturally occurring heavy chain antibody; (2) by expression of a nucleotide sequence encoding a naturally occurring V HH domain; (3) by "humanization” (as described herein) of a naturally occurring V HH domain or by expression of a nucleic acid encoding a such humanized VHH domain; (4) by "camelization” (as described herein) of a naturally occurring V H domain from any animal species, and in particular a from species of mammal, such as from a human being, or by expression of a nucleic acid encoding such a camelized VH domain; (5) by "camelisation” of a "domain antibody” or “Dab” as described by Ward et al (supra), or by expression of a nucleic acid encoding such a camelized V H domain; (6) by using synthetic or semi-synthetic techniques for preparing
  • VHH sequences corresponds to the V HH domains of naturally occurring heavy chain antibodies directed against a metalloproteinase from the ADAM family.
  • VHH sequences can generally be generated or obtained by suitably immunizing a species of Camelid with a metalloproteinase from the ADAM family (i.e. so as to raise an immune response and/or heavy chain antibodies directed against a metalloproteinase from the ADAM family), by obtaining a suitable biological sample from said Camelid (such as a blood sample, serum sample or sample of B-cells), and by generating V HH sequences directed against a metalloproteinase from the ADAM family, starting from said sample, using any suitable technique known per se.
  • VHH domains against a metalloproteinase from the ADAM family can be obtained from naive libraries of Camelid V HH sequences, for example by screening such a library using a metalloproteinase from the ADAM family, or at least one part, fragment, antigenic determinant or epitope thereof using one or more screening techniques known per se.
  • libraries and techniques are for example described in WO 99/37681, WO 01/90190, WO 03/025020 and WO 03/035694.
  • improved synthetic or semi-synthetic libraries derived from naive V HH libraries may be used, such as VHH libraries obtained from naive VHH libraries by techniques such as random mutagenesis and/or CDR shuffling, as for example described in WO 00/43507.
  • the invention relates to a method for generating Nanobodies, that are directed against a metalloproteinase from the ADAM family.
  • said method at least comprises the steps of: a) providing a set, collection or library of Nanobody sequences; and b) screening said set, collection or library of Nanobody sequences for Nanobody sequences that can bind to and/or have affinity for a metalloproteinase from the ADAM family; and c) isolating the amino acid sequence(s) that can bind to and/or have affinity for a metalloproteinase from the ADAM family.
  • the set, collection or library of Nanobody sequences may be a naive set, collection or library of Nanobody sequences; a synthetic or semi-synthetic set, collection or library of Nanobody sequences; and/or a set, collection or library of Nanobody sequences that have been subjected to affinity maturation.
  • the set, collection or library of Nanobody sequences may be an immune set, collection or library of Nanobody sequences, and in particular an immune set, collection or library of V HH sequences, that have been derived from a species of Camelid that has been suitably immunized with a metalloproteinase from the ADAM family or with a suitable antigenic determinant based thereon or derived therefrom, such as an antigenic part, fragment, region, domain, loop or other epitope thereof.
  • said antigenic determinant may be an extracellular part, region, domain, loop or other extracellular epitope(s).
  • the set, collection or library of Nanobody or V HH sequences may be displayed on a phage, phagemid, ribosome or suitable micro-organism (such as yeast), such as to facilitate screening.
  • suitable methods, techniques and host organisms for displaying and screening (a set, collection or library of) Nanobody sequences will be clear to the person skilled in the art, for example on the basis of the further disclosure herein. Reference is also made to WO 03/054016 and to the review by Hoogenboom in Nature Biotechnology, 23, 9, 1105-1116 (2005).
  • the method for generating Nanobody sequences comprises at least the steps of: a) providing a collection or sample of cells derived from a species of Camelid that express immunoglobulin sequences; b) screening said collection or sample of cells for (i) cells that express an immunoglobulin sequence that can bind to and/or have affinity for a metalloproteinase from the ADAM family; and (ii) cells that express heavy chain antibodies, in which substeps (i) and (ii) can be performed essentially as a single screening step or in any suitable order as two separate screening steps, so as to provide at least one cell that expresses a heavy chain antibody that can bind to and/or has affinity for a metalloproteinase from the ADAM family; and c) either (i) isolating from said cell the VHH sequence present in said heavy chain antibody; or (ii) isolating from said cell a nucleic acid sequence that encodes the V HH sequence present in said heavy chain antibody, followed by expressing said V
  • the collection or sample of cells may for example be a collection or sample of B-cells.
  • the sample of cells may be derived from a Camelid that has been suitably immunized with a metalloproteinase from the ADAM family or a suitable antigenic determinant based thereon or derived therefrom, such as an antigenic part, fragment, region, domain, loop or other epitope thereof.
  • said antigenic determinant may be an extracellular part, region, domain, loop or other extracellular epitope(s).
  • step b) is preferably performed using a flow cytometry technique such as FACS.
  • FACS flow cytometry technique
  • the method for generating an amino acid sequence directed against a metalloproteinase from the ADAM family may comprise at least the steps of: a) providing a set, collection or library of nucleic acid sequences encoding heavy chain antibodies or Nanobody sequences; b) screening said set, collection or library of nucleic acid sequences for nucleic acid sequences that encode a heavy chain antibody or a Nanobody sequence that can bind to and/or has affinity for a metalloproteinase from the ADAM family; and c) isolating said nucleic acid sequence, followed by expressing the VHH sequence present in said heavy chain antibody or by expressing said Nanobody sequence, respectively.
  • the set, collection or library of nucleic acid sequences encoding heavy chain antibodies or Nanobody sequences may for example be a set, collection or library of nucleic acid sequences encoding a naive set, collection or library of heavy chain antibodies or V HH sequences; a set, collection or library of nucleic acid sequences encoding a synthetic or semi-synthetic set, collection or library of Nanobody sequences; and/or a set, collection or library of nucleic acid sequences encoding a set, collection or library of Nanobody sequences that have been subjected to affinity maturation.
  • the set, collection or library of amino acid sequences may be an immune set, collection or library of nucleic acid sequences encoding heavy chain antibodies or V HH sequences derived from a Camelid that has been suitably immunized with a metalloproteinase from the ADAM family or with a suitable antigenic determinant based thereon or derived therefrom, such as an antigenic part, fragment, region, domain, loop or other epitope thereof.
  • said antigenic determinant may be an extracellular part, region, domain, loop or other extracellular epitope(s).
  • the set, collection or library of nucleotide sequences may be displayed on a phage, phagemid, ribosome or suitable micro-organism (such as yeast), such as to facilitate screening.
  • suitable methods, techniques and host organisms for displaying and screening (a set, collection or library of) nucleotide sequences encoding amino acid sequences will be clear to the person skilled in the art, for example on the basis of the further disclosure herein. Reference is also made to WO 03/054016 and to the review by Hoogenboom in Nature Biotechnology, 23, 9, 1105-1116 (2005).
  • the screening step of the methods described herein can also be performed as a selection step.
  • screening can comprise selection, screening or any suitable combination of selection and/or screening techniques.
  • a set, collection or library of sequences it may contain any suitable number of sequences, such as 1, 2, 3 or about 5, 10, 50, 100, 500, 1000, 5000, 10 4 , 10 5 , 10 6 , 10 7 , 10 8 or more sequences.
  • sequences in the above set, collection or library of amino acid sequences may be obtained or defined by rational, or semi-empirical approaches such as computer modelling techniques or biostatics or datamining techniques.
  • a set, collection or library can comprise one, two or more sequences that are variants from one another (e.g. with designed point mutations or with randomized positions), compromise multiple sequences derived from a diverse set of naturally diversified sequences (e.g. an immune library)), or any other source of diverse sequences (as described for example in Hoogenboom et al, Nat Biotechnol 23:1105, 2005 and Binz et al, Nat Biotechnol 2005, 23:1247).
  • Such set, collection or library of sequences can be displayed on the surface of a phage particle, a ribosome, a bacterium, a yeast cell, a mammalian cell, and linked to the nucleotide sequence encoding the amino acid sequence within these carriers.
  • a sequence is displayed on a suitable host or host cell, it is also possible (and customary) to first isolate from said host or host cell a nucleotide sequence that encodes the desired sequence, and then to obtain the desired sequence by suitably expressing said nucleotide sequence in a suitable host organism.
  • VHH sequences or Nanobody sequences directed against a metalloproteinase from the ADAM family involves suitably immunizing a transgenic mammal that is capable of expressing heavy chain antibodies (i.e.
  • V HH sequences or Nanobody sequences such as a blood sample, serum sample or sample of B-cells
  • VHH sequences directed against a metalloproteinase from the ADAM family starting from said sample, using any suitable technique known per se (such as any of the methods described herein or a hybridoma technique).
  • heavy chain antibody-expressing mice and the further methods and techniques described in WO 02/085945, WO 04/049794 and WO 06/008548 and Janssens et al, Proc. Natl. Acad. Sci .USA. 2006 Oct 10;103(41):15130-5 can be used.
  • heavy chain antibody expressing mice can express heavy chain antibodies with any suitable (single) variable domain, such as (single) variable domains from natural sources (e.g. human (single) variable domains, Camelid (single) variable domains or shark (single) variable domains), as well as for example synthetic or semi-synthetic (single) variable domains.
  • the invention also relates to the V HH sequences or Nanobody sequences that are obtained by the above methods, or alternatively by a method that comprises the one of the above methods and in addition at least the steps of determining the nucleotide sequence or amino acid sequence of said V HH sequence or Nanobody sequence; and of expressing or synthesizing said V HH sequence or Nanobody sequence in a manner known per se, such as by expression in a suitable host cell or host organism or by chemical synthesis.
  • a particularly preferred class of Nanobodies of the invention comprises Nanobodies with an amino acid sequence that corresponds to the amino acid sequence of a naturally occurring V HH domain, but that has been "humanized", i.e.
  • Nanobodies of the invention can be obtained in any suitable manner known per se (i.e. as indicated under points (1) - (8) above) and thus are not strictly limited to polypeptides that have been obtained using a polypeptide that comprises a naturally occurring V HH domain as a starting material.
  • Nanobodies of the invention comprises Nanobodies with an amino acid sequence that corresponds to the amino acid sequence of a naturally occurring V H domain, but that has been "camelized", i.e. by replacing one or more amino acid residues in the amino acid sequence of a naturally occurring VH domain from a conventional 4-chain antibody by one or more of the amino acid residues that occur at the corresponding position(s) in a V HH domain of a heavy chain antibody.
  • This can be performed in a manner known per se, which will be clear to the skilled person, for example on the basis of the further description herein.
  • the VH sequence that is used as a starting material or starting point for generating or designing the camelized Nanobody is preferably a V H sequence from a mammal, more preferably the V H sequence of a human being, such as a VH3 sequence.
  • camelized Nanobodies of the invention can be obtained in any suitable manner known per se (i.e. as indicated under points (1) - (8) above) and thus are not strictly limited to polypeptides that have been obtained using a polypeptide that comprises a naturally occurring VH domain as a starting material.
  • both “humanization” and “camelization” can be performed by providing a nucleotide sequence that encodes a naturally occurring VHH domain or VH domain, respectively, and then changing, in a manner known per se, one or more codons in said nucleotide sequence in such a way that the new nucleotide sequence encodes a "humanized” or “camelized” Nanobody of the invention, respectively.
  • This nucleic acid can then be expressed in a manner known per se, so as to provide the desired Nanobody of the invention.
  • the amino acid sequence of the desired humanized or camelized Nanobody of the invention can be designed and then synthesized de novo using techniques for peptide synthesis known per se.
  • a nucleotide sequence encoding the desired humanized or camelized Nanobody of the invention can be designed and then synthesized de novo using techniques for nucleic acid synthesis known per se, after which the nucleic acid thus obtained can be expressed in a manner known per se, so as to provide the desired Nanobody of the invention.
  • Nanobodies of the invention and/or nucleic acids encoding the same starting from naturally occurring V H sequences or preferably VHH sequences, will be clear from the skilled person, and may for example comprise combining one or more parts of one or more naturally occurring V H sequences (such as one or more FR sequences and/or CDR sequences), one or more parts of one or more naturally occurring VHH sequences (such as one or more FR sequences or CDR sequences), and/or one or more synthetic or semi-synthetic sequences, in a suitable manner, so as to provide a Nanobody of the invention or a nucleotide sequence or nucleic acid encoding the same (which may then be suitably expressed).
  • V H sequences such as one or more FR sequences and/or CDR sequences
  • synthetic or semi-synthetic sequences such as one or more synthetic or semi-synthetic sequences
  • Nucleotide sequences encoding framework sequences of VHH sequences or Nanobodies will be clear to the skilled person based on the disclosure herein and/or the further prior art cited herein (and/or may alternatively be obtained by PCR starting from the nucleotide sequences obtained using the methods described herein) and may be suitably combined with nucleotide sequences that encode the desired CDR's (for example, by PCR assembly using overlapping primers), so as to provide a nucleic acid encoding a Nanobody of the invention.
  • Nanobodies may in particular be characterized by the presence of one or more ""Hallmark residues ' " (as described herein) in one or more of the framework sequences.
  • a Nanobody in its broadest sense can be generally defined as a polypeptide comprising: a) an amino acid sequence that is comprised of four framework regions/sequences interrupted by three complementarity determining regions/sequences, in which the amino acid residue at position 108 according to the Kabat numbering is Q; and/or: b) an amino acid sequence that is comprised of four framework regions/sequences interrupted by three complementarity determining regions/sequences, in which the amino acid residue at position 45 according to the Kabat numbering is a charged amino acid (as defined herein) or a cysteine residue, and position 44 is preferably an E; and/or: c) an amino acid sequence that is comprised of four framework regions/sequences interrupted by three complementarity determining regions/sequences, in which the amino acid residue at position 103 according to the Kabat numbering is chosen from the group consisting of P, R and S, and
  • FRl - CDRl - FR2 - CDR2 - FR3 - CDR3 - FR4 in which FRl to FR4 refer to framework regions 1 to 4, respectively, and in which CDRl to CDR3 refer to the complementarity determining regions 1 to 3, respectively, and in which a) the amino acid residue at position 108 according to the Kabat numbering is Q; and/or in which: b) the amino acid residue at position 45 according to the Kabat numbering is a charged amino acid or a cysteine and the amino acid residue at position 44 according to the Kabat numbering is preferably E; and/or in which: c) the amino acid residue at position 103 according to the Kabat numbering is chosen from the group consisting of P, R and S, and is in particular chosen from the group consisting of R and S; and in which: d) CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined
  • a Nanobody in its broadest sense can be generally defined as a polypeptide comprising: a) an amino acid sequence that is comprised of four framework regions/sequences interrupted by three complementarity determining regions/sequences, in which the amino acid residue at position 108 according to the Kabat numbering is Q; and/or: b) an amino acid sequence that is comprised of four framework regions/sequences interrupted by three complementarity determining regions/sequences, in which the amino acid residue at position 44 according to the Kabat numbering is E and in which the amino acid residue at position 45 according to the Kabat numbering is an R; and/or: c) an amino acid sequence that is comprised of four framework regions/sequences interrupted by three complementarity determining regions/sequences, in which the amino acid residue at position 103 according to the Kabat numbering is chosen from the group consisting of P, R and S, and is in particular chosen from the group consisting ofR and S.
  • FRl to FR4 refer to framework regions 1 to 4, respectively, and in which CDRl to CDR3 refer to the complementarity determining regions 1 to 3, respectively, and in which a) the amino acid residue at position 108 according to the Kabat numbering is Q; and/or in which: b) the amino acid residue at position 44 according to the Kabat numbering is E and in which the amino acid residue at position 45 according to the Kabat numbering is an R; and/or in which: c) the amino acid residue at position 103 according to the Kabat numbering is chosen from the group consisting of P, R and S, and is in particular chosen from the group consisting of R and S ; and in which: d) CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • a Nanobody against a metalloproteinase from the ADAM family according to the invention may have the structure:
  • FRl to FR4 refer to framework regions 1 to 4, respectively, and in which CDRl to CDR3 refer to the complementarity determining regions 1 to 3, respectively, and in which a) the amino acid residue at position 108 according to the Kabat numbering is Q; and/or in which: b) the amino acid residue at position 44 according to the Kabat numbering is E and in which the amino acid residue at position 45 according to the Kabat numbering is an R; and/or in which: c) the amino acid residue at position 103 according to the Kabat numbering is chosen from the group consisting of P, R and S, and is in particular chosen from the group consisting of R and S; and in which: d) CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • a Nanobody can generally be defined as a polypeptide comprising an amino acid sequence that is comprised of four framework regions/sequences interrupted by three complementarity determining regions/sequences, in which; a-1) the amino acid residue at position 44 according to the Kabat numbering is chosen from the group consisting of A, G, E, D, G, Q, R, S, L; and is preferably chosen from the group consisting of G, E or Q; and a-2) the amino acid residue at position 45 according to the Kabat numbering is chosen from the group consisting of L, R or C; and is preferably chosen from the group consisting of L or R; and a-3) the amino acid residue at position 103 according to the Kabat numbering is chosen from the group consisting of W, R or S; and is preferably W or R, and is most preferably W; a-4) the amino acid residue at position 108 according to the Kabat numbering is Q; or in which:
  • a Nanobody of the invention may have the structure
  • FRl to FR4 refer to framework regions 1 to 4, respectively, and in which CDRl to CDR3 refer to the complementarity determining regions 1 to 3, respectively, and in which: a-1) the amino acid residue at position 44 according to the Kabat numbering is chosen from the group consisting of A, G, E, D, G, Q, R, S, L; and is preferably chosen from the group consisting of G, E or Q; and in which: a-2) the amino acid residue at position 45 according to the Kabat numbering is chosen from the group consisting of L, R or C; and is preferably chosen from the group consisting of
  • the amino acid residue at position 103 according to the Kabat numbering is chosen from the group consisting of W, R or S; and is preferably W or R, and is most preferably W; and in which a-4) the amino acid residue at position 108 according to the Kabat numbering is Q; and in which: d) CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • Nanobody of the invention may have the structure
  • FRl to FR4 refer to framework regions 1 to 4, respectively, and in which CDRl to CDR3 refer to the complementarity determining regions 1 to 3, respectively, and in which: b-1) the amino acid residue at position 44 according to the Kabat numbering is chosen from the group consisting of E and Q; and in which: b-2) the amino acid residue at position 45 according to the Kabat numbering is R; and in which: b-3) the amino acid residue at position 103 according to the Kabat numbering is chosen from the group consisting of W, R and S; and is preferably W; and in which: b-4) the amino acid residue at position 108 according to the Kabat numbering is chosen from the group consisting of Q and L; and is preferably Q; and in which: d) CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • FRl to FR4 refer to framework regions 1 to 4, respectively, and in which CDRl to CDR3 refer to the complementarity determining regions 1 to 3, respectively, and in which: c-1) the amino acid residue at position 44 according to the Kabat numbering is chosen from the group consisting of A, G, E, D, Q, R, S and L; and is preferably chosen from the group consisting of G, E and Q; and in which: c-2) the amino acid residue at position 45 according to the Kabat numbering is chosen from the group consisting of L, R and C; and is preferably chosen from the group consisting of L and R; and in which: c-3) the amino acid residue at position 103 according to the Kabat numbering is chosen from the group consisting of P, R and S; and is in particular chosen from the group consisting of R and S; and in which: c-4) the amino acid residue at position 108 according to the Kabat numbering is chosen from the group consisting of Q and L; is preferably Q; and in which: d)
  • Nanobodies of the invention are those according to a) above; according to (a-1) to (a-4) above; according to b) above; according to (b-1) to (b-4) above; according to (c) above; and/or according to (c-1) to (c-4) above, in which either: i) the amino acid residues at positions 44-47 according to the Kabat numbering form the sequence GLEW (or a GLEW-like sequence as described herein) and the amino acid residue at position 108 is Q; or in which: ii) the amino acid residues at positions 43-46 according to the Kabat numbering form the sequence KERE or KQRE (or a KERE -like sequence as described) and the amino acid residue at position 108 is Q or L, and is preferably Q.
  • a Nanobody of the invention may have the structure
  • FRl - CDRl - FR2 - CDR2 - FR3 - CDR3 - FR4 in which FRl to FR4 refer to framework regions 1 to 4, respectively, and in which CDRl to CDR3 refer to the complementarity determining regions 1 to 3, respectively, and in which: i) the amino acid residues at positions 44-47 according to the Kabat numbering form the sequence GLEW (or a GLEW-like sequence as defined herein) and the amino acid residue at position 108 is Q; and in which: ii) CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • Nanobody of the invention may have the structure
  • FRl to FR4 refer to framework regions 1 to 4, respectively, and in which CDRl to CDR3 refer to the complementarity determining regions 1 to 3, respectively, and in which: i) the amino acid residues at positions 43-46 according to the Kabat numbering form the sequence KERE or KQRE (or a KERE -like sequence) and the amino acid residue at position 108 is Q or L, and is preferably Q; and in which: ii) CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • the amino acid residues at positions 43-46 refer to framework regions 1 to 4, respectively, and in which CDRl to CDR3 refer to the complementarity determining regions 1 to 3, respectively, and in which: i) the amino acid residues at positions 43-46 according to the Kabat numbering form the sequence KERE or KQRE (or a KERE -like
  • the amino acid residue at position 37 is most preferably F.
  • the amino acid residue at position 37 is chosen from the group consisting of Y, H, I, L, V or F, and is most preferably V.
  • the Nanobodies of the invention can generally be classified on the basis of the following three groups: i) The "GLEW-group": Nanobodies with the amino acid sequence GLEW at positions 44- 47 according to the Kabat numbering and Q at position 108 according to the Kabat numbering. As further described herein, Nanobodies within this group usually have a V at position 37, and can have a W, P, R or S at position 103, and preferably have a W at position 103.
  • the GLEW group also comprises some GLEW-like sequences such as those mentioned in Table A-3 below.
  • Nanobodies belonging to the GLEW-group can be defined as Nanobodies with a G at position 44 and/or with a W at position 47, in which position 46 is usually E and in which preferably position 45 is not a charged amino acid residue and not cysteine; ii)
  • the "KERE-group” Nanobodies with the amino acid sequence KERE or KQRE (or another KERE-like sequence) at positions 43-46 according to the Kabat numbering and Q or L at position 108 according to the Kabat numbering.
  • Nanobodies within this group usually have a F at position 37, an L or F at position 47; and can have a W, P, R or S at position 103, and preferably have a W at position 103.
  • Nanobodies belonging to the KERE-group can be defined as Nanobodies with a K, Q or R at position 44 (usually K) in which position 45 is a charged amino acid residue or cysteine, and position 47 is as further defined herein; iii) The "703 P, R, S-group": Nanobodies with a P, R or S at position 103.
  • Nanobodies can have either the amino acid sequence GLEW at positions 44-47 according to the Kabat numbering or the amino acid sequence KERE or KQRE at positions 43-46 according to the Kabat numbering, the latter most preferably in combination with an F at position 37 and an L or an F at position 47 (as defined for the KERE-group); and can have Q or L at position 108 according to the Kabat numbering, and preferably have Q.
  • Nanobodies may belong to (i.e. have characteristics of) two or more of these classes.
  • one specifically preferred group of Nanobodies has GLEW or a GLEW-like sequence at positions 44-47; P, R or S (and in particular R) at position 103; and Q at position 108 (which may be humanized to L).
  • GLEW or GLEW-like sequence at positions 44-47; P, R or S (and in particular R) at position 103; and Q at position 108 (which may be humanized to L).
  • the definitions referred to above describe and apply to Nanobodies in the form of a native (i.e. non-humanized) VHH sequence, and that humanized variants of these Nanobodies may contain other amino acid residues than those indicated above (i.e. one or more humanizing substitutions as defined herein).
  • Nanobodies of the GLEW-group or the 103 P, R, S-group, Q at position 108 may be humanized to 108L.
  • other humanizing substitutions and suitable combinations thereof will become clear to the skilled person based on the disclosure herein.
  • a Nanobody of the invention may be a Nanobody belonging to the GLEW-group (as defined herein), and in which CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • a Nanobody of the invention may be a Nanobody belonging to the KERE-group (as defined herein), and CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • a Nanobody of the invention may be a Nanobody belonging to the 103 P, R, S-group (as defined herein), and in which CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • the Nanobodies of the invention can contain, at one or more positions that in a conventional V H domain would form (part of) the V H /V L interface, one or more amino acid residues that are more highly charged than the amino acid residues that naturally occur at the same position(s) in the corresponding naturally occurring V H sequence, and in particular one or more charged amino acid residues (as mentioned in Table A-2).
  • substitutions include, but are not limited to, the GLEW-like sequences mentioned in Table A-3 below; as well as the substitutions that are described in the International Application WO 00/29004 for so-called "microbodies", e.g. so as to obtain a Nanobody with Q at position 108 in combination with KLEW at positions 44-47 ' .
  • Other possible substitutions at these positions will be clear to the skilled person based upon the disclosure herein.
  • the amino acid residue at position 83 is chosen from the group consisting of L, M, S, V and W; and is preferably L.
  • the amino acid residue at position 83 is chosen from the group consisting of R, K, N, E, G, I, T and Q; and is most preferably either K or E (for Nanobodies corresponding to naturally occurring V HH domains) or R (for "humanized” Nanobodies, as described herein).
  • the amino acid residue at position 84 is chosen from the group consisting of P, A, R, S, D T, and V in one aspect, and is most preferably P (for Nanobodies corresponding to naturally occurring V HH domains) or R (for "humanized” Nanobodies, as described herein).
  • the amino acid residue at position 104 is chosen from the group consisting of G and D; and is most preferably G.
  • the amino acid residues at positions 11, 37, 44, 45, 47, 83, 84, 103, 104 and 108, which in the Nanobodies are as mentioned above, will also be referred to herein as the "Hallmark Residues".
  • the Hallmark Residues and the amino acid residues at the corresponding positions of the most closely related human V H domain, V H 3, are summarized in Table A-3. Some especially preferred but non-limiting combinations of these Hallmark Residues as occur in naturally occurring V HH domains are mentioned in Table A -4.
  • the corresponding amino acid residues of the human V H 3 called DP-47 have been indicated in italics.
  • Table A-4 Some preferred but non-limiting combinations of Hallmark Residues in naturally occurring Nanobodies.
  • each amino acid residue at any other position than the Hallmark Residues can be any amino acid residue that naturally occurs at the corresponding position (according to the Kabat numbering) of a naturally occurring V HH domain.
  • Tables A-5-A-8 also contain data on the VHH entropy ("VHH EnL”) and VHH variability ("VHH Var ”) at each amino acid position for a representative sample of 1118 V HH sequences (data kindly provided by David Lutje Hulsing and Prof. Theo Verrips of Utrecht University).
  • VHH EnL VHH entropy
  • VHH Var VHH variability
  • the values for the V HH entropy and the V HH variability provide a measure for the variability and degree of conservation of amino acid residues between the 1118 V HH sequences analyzed: low values (i.e. ⁇ 1, such as ⁇ 0.5) indicate that an amino acid residue is highly conserved between the V HH sequences (i.e. little variability).
  • the G at position 8 and the G at position 9 have values for the VHH entropy of 0.1 and 0 respectively, indicating that these residues are highly conserved and have little variability (and in case of position 9 is G in all 1118 sequences analysed), whereas for residues that form part of the CDR' s generally values of 1.5 or more are found (data not shown).
  • Table A-5 Non-limiting examples of amino acid residues in FRl (for the footnotes, see the footnotes to Table A-3)
  • Table A-6 Non-limiting examples of amino acid residues in FR2 (for the footnotes see the footnotes to Table A-3)
  • aNanobody of the invention can be defined as an amino acid sequence with the (general) structure
  • FRl to FR4 refer to framework regions 1 to 4, respectively, and in which CDRl to CDR3 refer to the complementarity determining regions 1 to 3, respectively, and in which: i) one or more of the amino acid residues at positions 11, 37, 44, 45, 47, 83, 84, 103, 104 and 108 according to the Kabat numbering are chosen from the Hallmark residues mentioned in Table A-3; and in which: ii) CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • Nanobodies may for example be V HH sequences or may be humanized Nanobodies.
  • VHH sequences they may be suitably humanized, as further described herein.
  • the Nanobodies are partially humanized Nanobodies, they may optionally be further suitably humanized, again as described herein.
  • aNanobody of the invention can be an amino acid sequence with the (general) structure
  • FRl to FR4 refer to framework regions 1 to 4, respectively, and in which CDRl to CDR3 refer to the complementarity determining regions 1 to 3, respectively, and in which: i) (preferably) one or more of the amino acid residues at positions 11, 37, 44, 45, 47, 83, 84, 103, 104 and 108 according to the Kabat numbering are chosen from the Hallmark residues mentioned in Table A-3 (it being understood that VHH sequences will contain one or more Hallmark residues; and that partially humanized Nanobodies will usually, and preferably, [still] contain one or more Hallmark residues [although it is also within the scope of the invention to provide - where suitable in accordance with the invention - partially humanized Nanobodies in which all Hallmark residues, but not one or more of the other amino acid residues, have been humanized]; and that in fully humanized Nanobodies, where suitable in accordance with the invention, all amino acid residues at the positions of the Hallmark residues will be amino acid residues that occur in a human V
  • V HH sequences such partially humanized Nanobodies with at least one Hallmark residue, such partially humanized Nanobodies without Hallmark residues and such fully humanized Nanobodies all form aspects of this invention); and in which: ii) said amino acid sequence has at least 80% amino acid identity with at least one of the amino acid sequences of SEQ ID NO's: 1 to 22, in which for the purposes of determining the degree of amino acid identity, the amino acid residues that form the CDR sequences (indicated with X in the sequences of SEQ ID NO's: 1 to 22) are disregarded; and in which: iii) CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • Nanobodies may for example be VHH sequences or may be humanized Nanobodies.
  • the above Nanobody sequences are V HH sequences, they may be suitably humanized, as further described herein.
  • the Nanobodies are partially humanized
  • Nanobodies they may optionally be further suitably humanized, again as described herein.
  • Table A-9 Representative amino acid sequences for Nanobodies of the KERE, GLEW and P,R,S 103 group.
  • Nanobody of the invention of the KERE group can be an amino acid sequence with the (general) structure
  • FRl - CDRl - FR2 - CDR2 - FR3 - CDR3 - FR4 in which: i) the amino acid residue at position 45 according to the Kabat numbering is a charged amino acid (as defined herein) or a cysteine residue, and position 44 is preferably an E; and in which: ii) FRl is an amino acid sequence that has at least 80% amino acid identity with at least one of the following amino acid sequences:
  • Table A-IO Representative FWl sequences for Nanobodies of the KERE-group.
  • FR2 is an amino acid sequence that has at least 80% amino acid identity with at least one of the following amino acid sequences:
  • Table A-Il Representative FW2 sequences for Nanobodies of the KERE-group.
  • FR3 is an amino acid sequence that has at least 80% amino acid identity with at least one of the following amino acid sequences:
  • Table A-12 Representative FW3 sequences for Nanobodies of the KERE-group.
  • FR4 is an amino acid sequence that has at least 80% amino acid identity with at least one of the following amino acid sequences: Table A-13: Representative FW4 sequences for Nanobodies of the KERE-group.
  • CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • one or more of the further Hallmark residues are preferably as described herein (for example, when they are V HH sequences or partially humanized Nanobodies).
  • the above Nanobodies may for example be VHH sequences or may be humanized Nanobodies.
  • the above Nanobody sequences are VHH sequences, they may be suitably humanized, as further described herein.
  • the Nanobodies are partially humanized Nanobodies, they may optionally be further suitably humanized, again as described herein.
  • framework 1 it will be clear to the skilled person that, when an amino acid sequence as outlined above is generated by expression of a nucleotide sequence, the first four amino acid sequences (i.e. amino acid residues 1-4 according to the Kabat numbering) may often be determined by the primer(s) that have been used to generate said nucleic acid. Thus, for determining the degree of amino acid identity, the first four amino acid residues are preferably disregarded.
  • amino acid positions 27 to 30 are according to the Kabat numbering considered to be part of the framework regions (and not the CDR's), it has been found by analysis of a database of more than 1000 V HH sequences that the positions 27 to 30 have a variability (expressed in terms of VHH entropy and VHH variability - see Tables A-5 to A-8) that is much greater than the variability on positions 1 to 26. Because of this, for determining the degree of amino acid identity, the amino acid residues at positions 27 to 30 are preferably also disregarded.
  • a Nanobody of the KERE class may be an amino acid sequence that is comprised of four framework regions/sequences interrupted by three complementarity determining regions/sequences, in which: i) the amino acid residue at position 45 according to the Kabat numbering is a charged amino acid (as defined herein) or a cysteine residue, and position 44 is preferably an E; and in which: ii) FRl is an amino acid sequence that, on positions 5 to 26 of the Kabat numbering, has at least 80% amino acid identity with at least one of the following amino acid sequences:
  • Table A-14 Representative FWl sequences (amino acid residues 5 to 26) for Nanobodies of the KERE-group.
  • FR2, FR3 and FR4 are as mentioned herein for FR2, FR3 and FR4 of Nanobodies of the KERE-class; and in which: iv) CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • the above Nanobodies may for example be V HH sequences or may be humanized Nanobodies.
  • VHH sequences they may be suitably humanized, as further described herein.
  • the Nanobodies are partially humanized Nanobodies, they may optionally be further suitably humanized, again as described herein.
  • a Nanobody of the GLEW class may be an amino acid sequence that is comprised of four framework regions/sequences interrupted by three complementarity determining regions/sequences, in which i) preferably, when the Nanobody of the GLEW-class is a non-humanized Nanobody, the amino acid residue in position 108 is Q; ii) FRl is an amino acid sequence that has at least 80% amino acid identity with at least one of the following amino acid sequences: Table A-15: Representative FWl sequences for Nanobodies of the GLEW-group.
  • FR2 is an amino acid sequence that has at least 80% amino acid identity with at least one of the following amino acid sequences:
  • Table A-16 Representative FW2 sequences for Nanobodies of the GLEW-group.
  • FR3 is an amino acid sequence that has at least 80% amino acid identity with at least one of the following amino acid sequences:
  • Table A-17 Representative FW3 sequences for Nanobodies of the GLEW-group.
  • FR4 is an amino acid sequence that has at least 80% amino acid identity with at least one of the following amino acid sequences:
  • Table A-18 Representative FW4 sequences for Nanobodies of the GLEW-group.
  • CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • one or more of the further Hallmark residues are preferably as described herein (for example, when they are VHH sequences or partially humanized Nanobodies).
  • a Nanobody of the GLEW class may be an amino acid sequence that is comprised of four framework regions/sequences interrupted by three complementarity determining regions/sequences, in which: i) preferably, when the Nanobody of the GLEW-class is a non-humanized Nanobody, the amino acid residue in position 108 is Q; and in which: ii) FRl is an amino acid sequence that, on positions 5 to 26 of the Kabat numbering, has at least 80% amino acid identity with at least one of the following amino acid sequences:
  • Table A-19 Representative FWl sequences (amino acid residues 5 to 26) for Nanobodies of the KERE-group.
  • FR2, FR3 and FR4 are as mentioned herein for FR2, FR3 and FR4 of Nanobodies of the GLEW-class; and in which: iv) CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • the above Nanobodies may for example be VHH sequences or may be humanized Nanobodies.
  • the above Nanobody sequences are V HH sequences, they may be suitably humanized, as further described herein.
  • the Nanobodies are partially humanized Nanobodies, they may optionally be further suitably humanized, again as described herein.
  • one or more of the further Hallmark residues are preferably as described herein (for example, when they are V HH sequences or partially humanized Nanobodies).
  • a Nanobody of the P, R, S 103 class may be an amino acid sequence that is comprised of four framework regions/sequences interrupted by three complementarity determining regions/sequences, in which i) the amino acid residue at position 103 according to the Kabat numbering is different from W; and in which: ii) preferably the amino acid residue at position 103 according to the Kabat numbering is
  • FRl is an amino acid sequence that has at least 80% amino acid identity with at least one of the following amino acid sequences:
  • Table A-20 Representative FWl sequences for Nanobodies of the P,R,S 103-group.
  • FR2 is an amino acid sequence that has at least 80% amino acid identity with at least one of the following amino acid sequences:
  • Table A-21 Representative FW2 sequences for Nanobodies of the P,R,S 103-group.
  • FR3 is an amino acid sequence that has at least 80% amino acid identity with at least one of the following amino acid sequences: Table A-22: Representative FW3 sequences for Nanobodies of the P,R,S 103-group.
  • FR4 is an amino acid sequence that has at least 80% amino acid identity with at least one of the following amino acid sequences:
  • Table A-23 Representative FW4 sequences for Nanobodies of the P,R,S 103-group.
  • CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • one or more of the further Hallmark residues are preferably as described herein (for example, when they are VHH sequences or partially humanized Nanobodies).
  • a Nanobody of the P,R,S 103 class may be an amino acid sequence that is comprised of four framework regions/sequences interrupted by three complementarity determining regions/sequences, in which: i) the amino acid residue at position 103 according to the Kabat numbering is different from W; and in which: ii) preferably the amino acid residue at position 103 according to the Kabat numbering is
  • FRl is an amino acid sequence that, on positions 5 to 26 of the Kabat numbering, has at least 80% amino acid identity with at least one of the following amino acid sequences:
  • Table A-24 Representative FWl sequences (amino acid residues 5 to 26) for Nanobodies of the P,R,S 103-group.
  • FR2, FR3 and FR4 are as mentioned herein for FR2, FR3 and FR4 of Nanobodies of the P,R,S 103 class; and in which: v) CDRl, CDR2 and CDR3 are as defined herein, and are preferably as defined according to one of the preferred aspects herein, and are more preferably as defined according to one of the more preferred aspects herein.
  • Nanobodies may for example be VHH sequences or may be humanized Nanobodies.
  • the above Nanobody sequences are V HH sequences, they may be suitably humanized, as further described herein.
  • the Nanobodies are partially humanized
  • Nanobodies they may optionally be further suitably humanized, again as described herein.
  • the invention relates to a Nanobody as described above, in which the CDR sequences have at least 70% amino acid identity, preferably at least 80% amino acid identity, more preferably at least 90% amino acid identity, such as 95% amino acid identity or more or even essentially 100% amino acid identity with the CDR sequences of at least one of the amino acid sequences of SEQ ID NO's: 868 to 973 and/or 1044 to 1053.
  • This degree of amino acid identity can for example be determined by determining the degree of amino acid identity (in a manner described herein) between said Nanobody and one or more of the sequences of SEQ ID NO's: 868 to 973 and/or 1044 to 1053, in which the amino acid residues that form the framework regions are disregarded.
  • Such Nanobodies can be as further described herein.
  • Nanobody with an amino acid sequence that is chosen from the group consisting of SEQ ID NO's: 868 to 973 and/or 1044 to 1053 or from the group consisting of from amino acid sequences that have more than 80%, preferably more than 90%, more preferably more than 95%, such as 99% or more sequence identity (as defined herein) with at least one of the amino acid sequences of SEQ ID NO's: 868 to 973 and/or 1044 to 1053.
  • any amino acid substitution (when it is not a humanizing substitution as defined herein) is preferably, and compared to the corresponding amino acid sequence of SEQ ID NO's: 868 to 973 and/or 1044 to 1053, a conservative amino acid substitution, (as defined herein); and/or: ii) its amino acid sequence preferably contains either only amino acid substitutions, or otherwise preferably no more than 5, preferably no more than 3, and more preferably only 1 or 2 amino acid deletions or insertions, compared to the corresponding amino acid sequence of SEQ ID NO's: 868 to 973 and/or 1044 to 1053; and/or iii) the CDR' s may be CDR' s that are derived by means of affinity maturation, for example starting from the CDR' s of to the corresponding amino acid sequence of SEQ ID NO's: 868 to 973 and/or 1044 to 1053.
  • the CDR sequences and FR sequences in the Nanobodies of the invention are such that the Nanobodies of the invention (and polypeptides of the invention comprising the same): bind to a metalloproteinase from the ADAM family with a dissociation constant (K D ) of 10 " to 10 " moles/liter or less, and preferably 10 " to 10 " moles/liter or less and more preferably 10 " to 10 " moles/liter (i.e.
  • K D dissociation constant
  • KA association
  • CDR sequences and FR sequences present in the Nanobodies of the invention are such that the Nanobodies of the invention will bind to a metalloproteinase from the ADAM family with an affinity less than 500 nM, preferably less than 200 nM, more preferably less than 10 nM, such as less than 500 pM.
  • a Nanobody may be as defined herein, but with the proviso that it has at least "one amino acid difference" (as defined herein) in at least one of the framework regions compared to the corresponding framework region of a naturally occurring human VH domain, and in particular compared to the corresponding framework region of DP-47.
  • a Nanobody may be as defined herein, but with the proviso that it has at least "one amino acid difference" (as defined herein) at at least one of the Hallmark residues (including those at positions 108, 103 and/or 45) compared to the corresponding framework region of a naturally occurring human V H domain, and in particular compared to the corresponding framework region of DP-47.
  • a Nanobody will have at least one such amino acid difference with a naturally occurring VH domain in at least one of FR2 and/or FR4, and in particular at at least one of the Hallmark residues in FR2 and/or FR4 (again, including those at positions 108, 103 and/or 45).
  • a humanized Nanobody of the invention may be as defined herein, but with the proviso that it has at least "one amino acid difference" (as defined herein) in at least one of the framework regions compared to the corresponding framework region of a naturally occurring V HH domain. More specifically, according to one non-limiting aspect of the invention, a humanized Nanobody may be as defined herein, but with the proviso that it has at least "one amino acid difference" (as defined herein) at at least one of the Hallmark residues (including those at positions 108, 103 and/or 45) compared to the corresponding framework region of a naturally occurring VHH domain.
  • a humanized Nanobody will have at least one such amino acid difference with a naturally occurring VHH domain in at least one of FR2 and/or FR4, and in particular at at least one of the Hallmark residues in FR2 and/or FR4 (again, including those at positions 108, 103 and/or 45).
  • Nanobodies of the invention As will be clear from the disclosure herein, it is also within the scope of the invention to use natural or synthetic analogs, mutants, variants, alleles, homologs and orthologs (herein collectively referred to as ""analogs") of the Nanobodies of the invention as defined herein, and in particular analogs of the Nanobodies of SEQ ID NO's 868 to 973 and/or 1044 to 1053.
  • the term "Nanobody of the invention” in its broadest sense also covers such analogs.
  • one or more amino acid residues may have been replaced, deleted and/or added, compared to the Nanobodies of the invention as defined herein.
  • Such substitutions, insertions or deletions may be made in one or more of the framework regions and/or in one or more of the CDR's.
  • substitutions, insertions or deletions are made in one or more of the framework regions, they may be made at one or more of the Hallmark residues and/or at one or more of the other positions in the framework residues, although substitutions, insertions or deletions at the Hallmark residues are generally less preferred (unless these are suitable humanizing substitutions as described herein).
  • a substitution may for example be a conservative substitution (as described herein) and/or an amino acid residue may be replaced by another amino acid residue that naturally occurs at the same position in another VHH domain (see Tables A-5 to A- 8 for some non-limiting examples of such substitutions), although the invention is generally not limited thereto.
  • any one or more substitutions, deletions or insertions, or any combination thereof, that either improve the properties of the Nanobody of the invention or that at least do not detract too much from the desired properties or from the balance or combination of desired properties of the Nanobody of the invention are included within the scope of the invention.
  • a skilled person will generally be able to determine and select suitable substitutions, deletions or insertions, or suitable combinations of thereof, based on the disclosure herein and optionally after a limited degree of routine experimentation, which may for example involve introducing a limited number of possible substitutions and determining their influence on the properties of the Nanobodies thus obtained.
  • deletions and/or substitutions may be designed in such a way that one or more sites for post-translational modification (such as one or more glycosylation sites) are removed, as will be within the ability of the person skilled in the art.
  • substitutions or insertions may be designed so as to introduce one or more sites for attachment of functional groups (as described herein), for example to allow site-specific pegylation (again as described herein).
  • the analogs are preferably such that they can bind to a metalloproteinase from the ADAM family with an affinity (suitably measured and/or expressed as a K D -value (actual or apparent), a KA-value (actual or apparent), a kon-rate and/or a k o ff-rate, or alternatively as an IC 5O value, as further described herein) that is as defined herein for the Nanobodies of the invention.
  • the analogs are preferably also such that they retain the favourable properties the Nanobodies, as described herein.
  • the analogs have a degree of sequence identity of at least 70%, preferably at least 80%, more preferably at least 90%, such as at least 95% or 99% or more; and/or preferably have at most 20, preferably at most 10, even more preferably at most 5, such as 4, 3, 2 or only 1 amino acid difference (as defined herein), with one of the Nanobodies of SEQ ID NOs: 868 to 973 and/or 1044 to 1053.
  • the framework sequences and CDR's of the analogs are preferably such that they are in accordance with the preferred aspects defined herein. More generally, as described herein, the analogs will have (a) a Q at position 108; and/or (b) a charged amino acid or a cysteine residue at position 45 and preferably an E at position 44, and more preferably E at position 44 and R at position 45; and/or (c) P, R or S at position 103.
  • Nanobodies of the invention comprise Nanobodies that have been humanized (i.e. compared to the sequence of a naturally occurring Nanobody of the invention).
  • humanization generally involves replacing one or more amino acid residues in the sequence of a naturally occurring V HH with the amino acid residues that occur at the same position in a human VH domain, such as a human VH3 domain.
  • Examples of possible humanizing substitutions or combinations of humanizing substitutions will be clear to the skilled person, for example from the Tables herein, from the possible humanizing substitutions mentioned in the background art cited herein, and/or from a comparison between the sequence of a Nanobody and the sequence of a naturally occurring human VH domain.
  • the humanizing substitutions should be chosen such that the resulting humanized Nanobodies still retain the favourable properties of Nanobodies as defined herein, and more preferably such that they are as described for analogs in the preceding paragraphs.
  • a skilled person will generally be able to determine and select suitable humanizing substitutions or suitable combinations of humanizing substitutions, based on the disclosure herein and optionally after a limited degree of routine experimentation, which may for example involve introducing a limited number of possible humanizing substitutions and determining their influence on the properties of the Nanobodies thus obtained.
  • the Nanobodies of the invention may become more "human-like", while still retaining the favorable properties of the Nanobodies of the invention as described herein.
  • Such humanized Nanobodies may have several advantages, such as a reduced immunogenicity, compared to the corresponding naturally occurring VHH domains.
  • the skilled person will be able to select humanizing substitutions or suitable combinations of humanizing substitutions which optimize or achieve a desired or suitable balance between the favourable properties provided by the humanizing substitutions on the one hand and the favourable properties of naturally occurring VHH domains on the other hand.
  • the Nanobodies of the invention may be suitably humanized at any framework residue(s), such as at one or more Hallmark residues (as defined herein) or at one or more other framework residues (i.e. non-Hallmark residues) or any suitable combination thereof.
  • Nanobodies of the "P,R,S-103 group” or the “KERE group” is Q 108 into L 108.
  • Nanobodies of the "GLEW class” may also be humanized by a Q108 into L108 substitution, provided at least one of the other Hallmark residues contains a camelid (camelizing) substitution (as defined herein).
  • one particularly preferred class of humanized Nanobodies has GLEW or a GLEW-like sequence at positions 44-47; P, R or S (and in particular R) at position 103, and an L at position 108.
  • the humanized and other analogs, and nucleic acid sequences encoding the same, can be provided in any manner known per se.
  • the analogs can be obtained by providing a nucleic acid that encodes a naturally occurring VHH domain, changing the codons for the one or more amino acid residues that are to be substituted into the codons for the corresponding desired amino acid residues (e.g. by site-directed mutagenesis or by PCR using suitable mismatch primers), expressing the nucleic acid/nucleotide sequence thus obtained in a suitable host or expression system; and optionally isolating and/or purifying the analog thus obtained to provide said analog in essentially isolated form (e.g. as further described herein).
  • nucleic acid encoding the desired analog can be synthesized in a manner known per se (for example using an automated apparatus for synthesizing nucleic acid sequences with a predefined amino acid sequence) and can then be expressed as described herein.
  • a technique may involve combining one or more naturally occurring and/or synthetic nucleic acid sequences each encoding a part of the desired analog, and then expressing the combined nucleic acid sequence as described herein.
  • the analogs can be provided using chemical synthesis of the pertinent amino acid sequence using techniques for peptide synthesis known per se, such as those mentioned herein.
  • the Nanobodies of the invention can be designed and/or prepared starting from human V H sequences (i.e. amino acid sequences or the corresponding nucleotide sequences), such as for example from human VH3 sequences such as DP-47, DP-51 or DP-29, i.e. by introducing one or more camelizing substitutions (i.e.
  • camelizing substitutions can be derived from Tables A-5 - A-8. It will also be clear that camelizing substitutions at one or more of the Hallmark residues will generally have a greater influence on the desired properties than substitutions at one or more of the other amino acid positions, although both and any suitable combination thereof are included within the scope of the invention. For example, it is possible to introduce one or more camelizing substitutions that already confer at least some the desired properties, and then to introduce further camelizing substitutions that either further improve said properties and/or confer additional favourable properties.
  • such camelizing substitutions are preferably such that the resulting an amino acid sequence at least contains (a) a Q at position 108; and/or (b) a charged amino acid or a cysteine residue at position 45 and preferably also an E at position 44, and more preferably E at position 44 and R at position 45; and/or (c) P, R or S at position 103; and optionally one or more further camelizing substitutions. More preferably, the camelizing substitutions are such that they result in a Nanobody of the invention and/or in an analog thereof (as defined herein), such as in a humanized analog and/or preferably in an analog that is as defined in the preceding paragraphs.
  • Nanobodies of SEQ ID NO's: 868 to 973 and/or 1044 to 1053 are nanobodies of SEQ ID NO's: 868 to 973 and/or 1044 to 1053.
  • the term "Nanobody of the invention” in its broadest sense also covers such parts or fragments.
  • such parts or fragments of the Nanobodies of the invention have amino acid sequences in which, compared to the amino acid sequence of the corresponding full length Nanobody of the invention (or analog thereof), one or more of the amino acid residues at the N-terminal end, one or more amino acid residues at the C- terminal end, one or more contiguous internal amino acid residues, or any combination thereof, have been deleted and/or removed.
  • the parts or fragments are preferably such that they can bind to a metalloproteinase from the ADAM family with an affinity (suitably measured and/or expressed as a K D -value (actual or apparent), a KA-value (actual or apparent), a k on -rate and/or a k o ff-rate, or alternatively as an IC50 value, as further described herein) that is as defined herein for the Nanobodies of the invention.
  • an affinity suitably measured and/or expressed as a K D -value (actual or apparent), a KA-value (actual or apparent), a k on -rate and/or a k o ff-rate, or alternatively as an IC50 value, as further described herein
  • any part or fragment is preferably such that it comprises at least 10 contiguous amino acid residues, preferably at least 20 contiguous amino acid residues, more preferably at least 30 contiguous amino acid residues, such as at least 40 contiguous amino acid residues, of the amino acid sequence of the corresponding full length Nanobody of the invention. Also, any part or fragment is such preferably that it comprises at least one of CDRl ,
  • any part or fragment is such that it comprises at least one of the CDR's (and preferably at least CDR3 or part thereof) and at least one other CDR (i.e. CDRl or CDR2) or at least part thereof, preferably connected by suitable framework sequence(s) or at least part thereof. More preferably, any part or fragment is such that it comprises at least one of the CDR's (and preferably at least CDR3 or part thereof) and at least part of the two remaining CDR's, again preferably connected by suitable framework sequence(s) or at least part thereof.
  • such a part or fragment comprises at least CDR3, such as FR3, CDR3 and FR4 of the corresponding full length Nanobody of the invention, i.e. as for example described in the International application WO 03/050531 (Lasters et al).
  • Nanobody of the invention it is also possible to combine two or more of such parts or fragments (i.e. from the same or different Nanobodies of the invention), i.e. to provide an analog (as defined herein) and/or to provide further parts or fragments (as defined herein) of a Nanobody of the invention. It is for example also possible to combine one or more parts or fragments of a Nanobody of the invention with one or more parts or fragments of a human VH domain.
  • the parts or fragments have a degree of sequence identity of at least 50%, preferably at least 60%, more preferably at least 70%, even more preferably at least 80%, such as at least 90%, 95% or 99% or more with one of the Nanobodies of SEQ ID NOs 868 to 973 and/or 1044 to 1053.
  • the parts and fragments, and nucleic acid sequences encoding the same can be provided and optionally combined in any manner known per se.
  • such parts or fragments can be obtained by inserting a stop codon in a nucleic acid that encodes a full-sized Nanobody of the invention, and then expressing the nucleic acid thus obtained in a manner known per se (e.g. as described herein).
  • nucleic acids encoding such parts or fragments can be obtained by suitably restricting a nucleic acid that encodes a full-sized Nanobody of the invention or by synthesizing such a nucleic acid in a manner known per se.
  • Parts or fragments may also be provided using techniques for peptide synthesis known per se.
  • the invention in its broadest sense also comprises derivatives of the Nanobodies of the invention.
  • derivatives can generally be obtained by modification, and in particular by chemical and/or biological (e.g. enzymatical) modification, of the Nanobodies of the invention and/or of one or more of the amino acid residues that form the Nanobodies of the invention.
  • such a modification may involve the introduction (e.g. by covalent linking or in an other suitable manner) of one or more functional groups, residues or moieties into or onto the Nanobody of the invention, and in particular of one or more functional groups, residues or moieties that confer one or more desired properties or functionalities to the Nanobody of the invention.
  • one or more functional groups, residues or moieties may be clear to the skilled person.
  • such modification may comprise the introduction (e.g. by covalent binding or in any other suitable manner) of one or more functional groups that increase the half-life, the solubility and/or the absorption of the Nanobody of the invention, that reduce the immunogenicity and/or the toxicity of the Nanobody of the invention, that eliminate or attenuate any undesirable side effects of the Nanobody of the invention, and/or that confer other advantageous properties to and/or reduce the undesired properties of the Nanobodies and/or polypeptides of the invention; or any combination of two or more of the foregoing.
  • Such functional groups can generally comprise all functional groups and techniques mentioned in the general background art cited hereinabove as well as the functional groups and techniques known per se for the modification of pharmaceutical proteins, and in particular for the modification of antibodies or antibody fragments (including ScFv's and single domain antibodies), for which reference is for example made to Remington's Pharmaceutical Sciences, 16th ed., Mack Publishing Co., Easton, PA (1980).
  • Such functional groups may for example be linked directly (for example covalently) to a Nanobody of the invention, or optionally via a suitable linker or spacer, as will again be clear to the skilled person.
  • One of the most widely used techniques for increasing the half-life and/or reducing the immunogenicity of pharmaceutical proteins comprises attachment of a suitable pharmacologically acceptable polymer, such as poly(ethyleneglycol) (PEG) or derivatives thereof (such as methoxypoly(ethyleneglycol) or mPEG).
  • PEG poly(ethyleneglycol)
  • derivatives thereof such as methoxypoly(ethyleneglycol) or mPEG.
  • pegylation can be used, such as the pegylation used in the art for antibodies and antibody fragments (including but not limited to (single) domain antibodies and ScFv's); reference is made to for example Chapman, Nat. BiotechnoL, 54, 531-545 (2002); by Veronese and Harris, Adv. Drug Deliv. Rev. 54, 453-456 (2003), by Harris and Chess, Nat. Rev. Drug.
  • site-directed pegylation is used, in particular via a cysteine-residue (see for example Yang et al., Protein Engineering, 16, 10, 761-770 (2003).
  • PEG may be attached to a cysteine residue that naturally occurs in a Nanobody of the invention
  • a Nanobody of the invention may be modified so as to suitably introduce one or more cysteine residues for attachment of PEG, or an amino acid sequence comprising one or more cysteine residues for attachment of PEG may be fused to the N- and/or C-terminus of a Nanobody of the invention, all using techniques of protein engineering known per se to the skilled person.
  • a PEG is used with a molecular weight of more than 5000, such as more than 10,000 and less than 200,000, such as less than 100,000; for example in the range of 20,000-80,000.
  • Another, usually less preferred modification comprises N-linked or O-linked glycosylation, usually as part of co-translational and/or post-translational modification, depending on the host cell used for expressing the Nanobody or polypeptide of the invention.
  • Yet another modification may comprise the introduction of one or more detectable labels or other signal-generating groups or moieties, depending on the intended use of the labelled Nanobody.
  • Suitable labels and techniques for attaching, using and detecting them will be clear to the skilled person, and for example include, but are not limited to, fluorescent labels (such as fluorescein, isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde, and fiuorescamine and fluorescent metals such as Eu or others metals from the lanthanide series), phosphorescent labels, chemiluminescent labels or bioluminescent labels (such as luminal, isoluminol, theromatic acridinium ester, imidazole, acridinium salts, oxalate ester, dioxetane or GFP and its analogs), radio-isotopes (such as 3 H, 125 1, 32 P, 35 S, 14 C
  • Nanobodies and polypeptides of the invention may for example be used for in vitro, in vivo or in situ assays (including immunoassays known per se such as ELISA, RIA, EIA and other "sandwich assays", etc.) as well as in vivo diagnostic and imaging purposes, depending on the choice of the specific label.
  • chelating group for example to chelate one of the metals or metallic cations referred to above.
  • Suitable chelating groups for example include, without limitation, diethyl- enetriaminepentaacetic acid (DTPA) or ethylenediaminetetraacetic acid (EDTA).
  • DTPA diethyl- enetriaminepentaacetic acid
  • EDTA ethylenediaminetetraacetic acid
  • Yet another modification may comprise the introduction of a functional group that is one part of a specific binding pair, such as the biotin-(strept)avidin binding pair.
  • a functional group may be used to link the Nanobody of the invention to another protein, polypeptide or chemical compound that is bound to the other half of the binding pair, i.e. through formation of the binding pair.
  • a Nanobody of the invention may be conjugated to biotin, and linked to another protein, polypeptide, compound or carrier conjugated to avidin or streptavidin.
  • such a conjugated Nanobody may be used as a reporter, for example in a diagnostic system where a detectable signal-producing agent is conjugated to avidin or streptavidin.
  • binding pairs may for example also be used to bind the Nanobody of the invention to a carrier, including carriers suitable for pharmaceutical purposes.
  • a carrier including carriers suitable for pharmaceutical purposes.
  • Such binding pairs may also be used to link a therapeutically active agent to the Nanobody of the invention.
  • the Nanobodies of the invention may also be linked to a toxin or to a toxic residue or moiety. Examples of toxic moieties, compounds or residues which can be linked to a
  • Nanobody of the invention to provide - for example - a cytotoxic compound will be clear to the skilled person and can for example be found in the prior art cited above and/or in the further description herein.
  • One example is the so-called ADEPTTM technology described in WO 03/055527.
  • Other potential chemical and enzymatical modifications will be clear to the skilled person. Such modifications may also be introduced for research purposes (e.g. to study function-activity relationships). Reference is for example made to Lundblad and Bradshaw, Biotechnol. Appl. Biochem., 26, 143-151 (1997).
  • the derivatives are such that they bind to a metalloproteinase from the ADAM family with an affinity (suitably measured and/or expressed as a K D -value (actual or apparent), a KA-value (actual or apparent), a kon-rate and/or a k o ff-rate, or alternatively as an IC 5O value, as further described herein) that is as defined herein for the Nanobodies of the invention.
  • the invention also relates to proteins or polypeptides that essentially consist of or comprise at least one Nanobody of the invention.
  • essentially consist of is meant that the amino acid sequence of the polypeptide of the invention either is exactly the same as the amino acid sequence of a Nanobody of the invention or corresponds to the amino acid sequence of a Nanobody of the invention which has a limited number of amino acid residues, such as 1-20 amino acid residues, for example 1-10 amino acid residues and preferably 1-6 amino acid residues, such as 1, 2, 3, 4, 5 or 6 amino acid residues, added at the amino terminal end, at the carboxy terminal end, or at both the amino terminal end and the carboxy terminal end of the amino acid sequence of the Nanobody.
  • amino acid residues may or may not change, alter or otherwise influence the (biological) properties of the Nanobody and may or may not add further functionality to the Nanobody.
  • amino acid residues can comprise an N-terminal Met residue, for example as result of expression in a heterologous host cell or host organism. may form a signal sequence or leader sequence that directs secretion of the Nanobody from a host cell upon synthesis. Suitable secretory leader peptides will be clear to the skilled person, and may be as further described herein.
  • such a leader sequence will be linked to the N-terminus of the Nanobody, although the invention in its broadest sense is not limited thereto; may form a sequence or signal that allows the Nanobody to be directed towards and/or to penetrate or enter into specific organs, tissues, cells, or parts or compartments of cells, and/or that allows the Nanobody to penetrate or cross a biological barrier such as a cell membrane, a cell layer such as a layer of epithelial cells, a tumor including solid tumors, or the blood-brain-barrier.
  • a biological barrier such as a cell membrane, a cell layer such as a layer of epithelial cells, a tumor including solid tumors, or the blood-brain-barrier.
  • Examples of such amino acid sequences will be clear to the skilled person.
  • Some non-limiting examples are the small peptide vectors ("Pep-trans vectors") described in WO 03/026700 and in Temsamani et al., Expert
  • Intrabodies comprising a Nanobody of the invention, as mentioned below; may form a "tag", for example an amino acid sequence or residue that allows or facilitates the purification of the Nanobody, for example using affinity techniques directed against said sequence or residue. Thereafter, said sequence or residue may be removed (e.g. by chemical or enzymatical cleavage) to provide the Nanobody sequence (for this purpose, the tag may optionally be linked to the Nanobody sequence via a cleavable linker sequence or contain a cleavable motif).
  • residues are multiple histidine residues, glutathione residues and a myc-tag (see for example SEQ ID NO:31 of WO 06/12282).
  • Suitable amino acid residues and functional groups will be clear to the skilled person and include, but are not limited to, the amino acid residues and functional groups mentioned herein for the derivatives of the Nanobodies of the invention.
  • a polypeptide of the invention comprises a Nanobody of the invention, which is fused at its amino terminal end, at its carboxy terminal end, or both at its amino terminal end and at its carboxy terminal end to at least one further amino acid sequence, i.e. so as to provide a fusion protein comprising said Nanobody of the invention and the one or more further amino acid sequences.
  • a fusion will also be referred to herein as a "Nanobody fusion".
  • the one or more further amino acid sequence may be any suitable and/or desired amino acid sequences.
  • the further amino acid sequences may or may not change, alter or otherwise influence the (biological) properties of the Nanobody, and may or may not add further functionality to the Nanobody or the polypeptide of the invention.
  • the further amino acid sequence is such that it confers one or more desired properties or functionalities to the Nanobody or the polypeptide of the invention.
  • the further amino acid sequence may also provide a second binding site, which binding site may be directed against any desired protein, polypeptide, antigen, antigenic determinant or epitope (including but not limited to the same protein, polypeptide, antigen, antigenic determinant or epitope against which the Nanobody of the invention is directed, or a different protein, polypeptide, antigen, antigenic determinant or epitope).
  • amino acid sequences will be clear to the skilled person, and may generally comprise all amino acid sequences that are used in peptide fusions based on conventional antibodies and fragments thereof (including but not limited to ScFv's and single domain antibodies). Reference is for example made to the review by Holliger and Hudson, Nature Biotechnology, 23, 9, 1126-1136 (2005),
  • such an amino acid sequence may be an amino acid sequence that increases the half-life, the solubility, or the absorption, reduces the immunogenicity or the toxicity, eliminates or attenuates undesirable side effects, and/or confers other advantageous properties to and/or reduces the undesired properties of the polypeptides of the invention, compared to the Nanobody of the invention per se.
  • Some non-limiting examples of such amino acid sequences are serum proteins, such as human serum albumin (see for example WO 00/27435) or haptenic molecules (for example haptens that are recognized by circulating antibodies, see for example WO 98/22141).
  • the Nanobody of the invention is preferably either directly linked to serum albumin (or to a suitable fragment thereof) or via a suitable linker, and in particular via a suitable peptide linked so that the polypeptide of the invention can be expressed as a genetic fusion (protein).
  • the Nanobody of the invention may be linked to a fragment of serum albumin that at least comprises the domain III of serum albumin or part thereof.
  • the further amino acid sequence may provide a second binding site or binding unit that is directed against a serum protein (such as, for example, human serum albumin or another serum protein such as IgG), so as to provide increased half-life in serum.
  • a serum protein such as, for example, human serum albumin or another serum protein such as IgG
  • amino acid sequences for example include the Nanobodies described below, as well as the small peptides and binding proteins described in WO 91/01743, WO 01/45746 and WO 02/076489 and the dAb's described in WO 03/002609 and WO 04/003019.
  • amino acid sequences may in particular be directed against serum albumin (and more in particular human serum albumin) and/or against IgG (and more in particular human IgG).
  • such amino acid sequences may be amino acid sequences that are directed against (human) serum albumin and amino acid sequences that can bind to amino acid residues on (human) serum albumin that are not involved in binding of serum albumin to FcRn (see for example WO 06/0122787) and/or amino acid sequences that are capable of binding to amino acid residues on serum albumin that do not form part of domain III of serum albumin (see again for example WO 06/0122787); amino acid sequences that have or can provide an increased half-life (see for example the US provisional application 60/843,349 by Ablynx N.V.
  • the one or more further amino acid sequences may comprise one or more parts, fragments or domains of conventional 4-chain antibodies (and in particular human antibodies) and/or of heavy chain antibodies.
  • aNanobody of the invention may be linked to a conventional (preferably human) VH or VL domain or to a natural or synthetic analog of a VH or VL domain, again optionally via a linker sequence (including but not limited to other (single) domain antibodies, such as the dAb's described by Ward et al.).
  • the at least one Nanobody may also be linked to one or more (preferably human)
  • CHI , CH2 and/or CH3 domains optionally via a linker sequence.
  • aNanobody linked to a suitable C H 1 domain could for example be used - together with suitable light chains - to generate antibody fragments/structures analogous to conventional Fab fragments or F(ab') 2 fragments, but in which one or (in case of an F(ab') 2 fragment) one or both of the conventional V H domains have been replaced by a Nanobody of the invention.
  • two Nanobodies could be linked to a C H 3 domain (optionally via a linker) to provide a construct with increased half-life in vivo.
  • one or more Nanobodies of the invention may be linked (optionally via a suitable linker or hinge region) to one or more constant domains (for example, 2 or 3 constant domains that can be used as part of/to form an Fc portion), to an Fc portion and/or to to one or more antibody parts, fragments or domains that confer one or more effector functions to the polypeptide of the invention and/or may confer the ability to bind to one or more Fc receptors.
  • constant domains for example, 2 or 3 constant domains that can be used as part of/to form an Fc portion
  • an Fc portion and/or to to one or more antibody parts, fragments or domains that confer one or more effector functions to the polypeptide of the invention and/or may confer the ability to bind to one or more Fc receptors.
  • the one or more further amino acid sequences may comprise one or more C H 2 and/or C H 3 domains of an antibody, such as from a heavy chain antibody (as described herein) and more preferably from a conventional human 4-chain antibody; and/or may form (part of) and Fc region, for example from IgG (e.g. from IgGl, IgG2, IgG3 or IgG4), from IgE or from another human Ig such as IgA, IgD or IgM.
  • WO 94/04678 describes heavy chain antibodies comprising a Camelid V HH domain or a humanized derivative thereof (i.e.
  • Nanobody in which the Camelidae C H 2 and/or CH3 domain have been replaced by human CH2 and CH3 domains, so as to provide an immunoglobulin that consists of 2 heavy chains each comprising a Nanobody and human C H 2 and C H 3 domains (but no C H 1 domain), which immunoglobulin has the effector function provided by the C H 2 and C H 3 domains and which immunoglobulin can function without the presence of any light chains.
  • Other amino acid sequences that can be suitably linked to the Nanobodies of the invention so as to provide an effector function will be clear to the skilled person, and may be chosen on the basis of the desired effector function(s).
  • C H 2 and/or C H 3 domains that confer increased half- life without any biologically significant effector function may also be suitable or even preferred.
  • suitable constructs comprising one or more Nanobodies and one or more constant domains with increased half-life in vivo will be clear to the skilled person, and may for example comprise two Nanobodies linked to a CH3 domain, optionally via a linker sequence.
  • any fusion protein or derivatives with increased half-life will preferably have a molecular weight of more than 50 kD, the cut-off value for renal absorption.
  • one or more amino acid sequences of the invention may be linked (optionally via a suitable linker or hinge region) to naturally occurring, synthetic or semisynthetic constant domains (or analogs, variants, mutants, parts or fragments thereof) that have a reduced (or essentially no) tendency to self-associate into dimers (i.e. compared to constant domains that naturally occur in conventional 4-chain antibodies).
  • Such monomeric (i.e. not self- associating) Fc chain variants, or fragments thereof will be clear to the skilled person. For example, Helm et al., J Biol Chem 1996 271 7494, describe monomeric Fc ⁇ chain variants that can be used in the polypeptide chains of the invention.
  • such monomeric Fc chain variants are preferably such that they are still capable of binding to the complement or the relevant Fc receptor(s) (depending on the Fc portion from which they are derived), and/or such that they still have some or all of the effector functions of the Fc portion from which they are derived (or at a reduced level still suitable for the intended use).
  • the monomeric Fc chain may be used to confer increased half-life upon the polypeptide chain, in which case the monomeric Fc chain may also have no or essentially no effector functions.
  • Bivalent/multivalent, bispecific/multispecific or biparatopic/multiparatopic polypeptides of the invention may also be linked to Fc portions, in order to provide polypeptide constructs of the type that is described in the non-prepublished US provisional application entitled “"immunoglobulin constructs” filed on December 4, 2007.
  • the further amino acid sequences may also form a signal sequence or leader sequence that directs secretion of the Nanobody or the polypeptide of the invention from a host cell upon synthesis (for example to provide a pre-, pro- or prepro- form of the polypeptide of the invention, depending on the host cell used to express the polypeptide of the invention).
  • the further amino acid sequence may also form a sequence or signal that allows the Nanobody or polypeptide of the invention to be directed towards and/or to penetrate or enter into specific organs, tissues, cells, or parts or compartments of cells, and/or that allows the Nanobody or polypeptide of the invention to penetrate or cross a biological barrier such as a cell membrane, a cell layer such as a layer of epithelial cells, a tumor including solid tumors, or the blood-brain-barrier.
  • Suitable examples of such amino acid sequences will be clear to the skilled person, and for example include, but are not limited to, the "Peptrans" vectors mentioned above, the sequences described by Cardinale et al.
  • Nanobodies and polypeptides of the invention as so-called “intrabodies”, for example as described in WO 94/02610, WO 95/22618, US-A-7004940, WO 03/014960, WO 99/07414; WO 05/01690; EP 1 512 696; and in Cattaneo, A. & Biocca, S. (1997) Intracellular Antibodies: Development and Applications. Austin and Springer- Verlag; and in Kontermann, Methods 34, (2004), 163-170, and the further references described therein.
  • the Nanobodies of the invention may also be linked to a (cyto)toxic protein or polypeptide.
  • ADEPTTM technology described in WO 03/055527.
  • said one or more further amino acid sequences comprise at least one further Nanobody, so as to provide a polypeptide of the invention that comprises at least two, such as three, four, five or more Nanobodies, in which said Nanobodies may optionally be linked via one or more linker sequences (as defined herein).
  • Polypeptides of the invention that comprise two or more Nanobodies, of which at least one is a Nanobody of the invention will also be referred to herein as "multivalent" polypeptides of the invention, and the Nanobodies present in such polypeptides will also be referred to herein as being in a "multivalent format".
  • a “bivalent” polypeptide of the invention comprises two Nanobodies, optionally linked via a linker sequence
  • a “trivalent” polypeptide of the invention comprises three Nanobodies, optionally linked via two linker sequences; etc.; in which at least one of the Nanobodies present in the polypeptide, and up to all of the Nanobodies present in the polypeptide, is/are a Nanobody of the invention.
  • the two or more Nanobodies may be the same or different, and may be directed against the same antigen or antigenic determinant (for example against the same part(s) or epitope(s) or against different parts or epitopes) or may alternatively be directed against different antigens or antigenic determinants; or any suitable combination thereof.
  • a bivalent polypeptide of the invention may comprise (a) two identical Nanobodies; (b) a first Nanobody directed against a first antigenic determinant of a protein or antigen and a second Nanobody directed against the same antigenic determinant of said protein or antigen which is different from the first Nanobody; (c) a first Nanobody directed against a first antigenic determinant of a protein or antigen and a second Nanobody directed against another antigenic determinant of said protein or antigen; or (d) a first Nanobody directed against a first protein or antigen and a second Nanobody directed against a second protein or antigen (i.e. different from said first antigen).
  • a trivalent polypeptide of the invention may, for example and without being limited thereto, comprise (a) three identical Nanobodies; (b) two identical Nanobody against a first antigenic determinant of an antigen and a third Nanobody directed against a different antigenic determinant of the same antigen; (c) two identical Nanobody against a first antigenic determinant of an antigen and a third Nanobody directed against a second antigen different from said first antigen; (d) a first Nanobody directed against a first antigenic determinant of a first antigen, a second Nanobody directed against a second antigenic determinant of said first antigen and a third Nanobody directed against a second antigen different from said first antigen; or (e) a first Nanobody directed against a first antigen, a second Nanobody directed against a second antigen different from said first antigen, and a third Nanobody directed against a third antigen different from said first and second antigen.
  • Polypeptides of the invention that contain at least two Nanobodies, in which at least one Nanobody is directed against a first antigen (i.e. against a metalloproteinase from the ADAM family,) and at least one Nanobody is directed against a second antigen (i.e. different from a metalloproteinase from the ADAM family,), will also be referred to as "multispecific" polypeptides of the invention, and the Nanobodies present in such polypeptides will also be referred to herein as being in a "multispecific format".
  • a "bispecific" polypeptide of the invention is a polypeptide that comprises at least one Nanobody directed against a first antigen (i.e.
  • a "trispecific" polypeptide of the invention is a polypeptide that comprises at least one Nanobody directed against a first antigen (i.e. a metalloproteinase from the ADAM family,), at least one further Nanobody directed against a second antigen (i.e. different from a metalloproteinase from the ADAM family,) and at least one further Nanobody directed against a third antigen (i.e. different from both a metalloproteinase from the ADAM family, and the second antigen); etc.
  • a bispecific polypeptide of the invention is a bivalent polypeptide of the invention (as defined herein), comprising a first Nanobody directed against a metalloproteinase from the ADAM family, and a second Nanobody directed against a second antigen, in which said first and second Nanobody may optionally be linked via a linker sequence (as defined herein);
  • a trispecific polypeptide of the invention in its simplest form is a trivalent polypeptide of the invention (as defined herein), comprising a first Nanobody directed against a metalloproteinase from the ADAM family, a second Nanobody directed against a second antigen and a third Nanobody directed against a third antigen, in which said first, second and third Nanobody may optionally be linked via one or more, and in particular one and more, in particular two, linker sequences.
  • a multispecific polypeptide of the invention may comprise at least one Nanobody against a metalloproteinase from the ADAM family, and any number of Nanobodies directed against one or more antigens different from a metalloproteinase from the ADAM family.
  • the specific order or arrangement of the various Nanobodies in the polypeptides of the invention may have some influence on the properties of the final polypeptide of the invention (including but not limited to the affinity, specificity or avidity for a metalloproteinase from the ADAM family, or against the one or more other antigens), said order or arrangement is usually not critical and may be suitably chosen by the skilled person, optionally after some limited routine experiments based on the disclosure herein.
  • a specific multivalent or multispecific polypeptide of the invention it should be noted that this encompasses any order or arrangements of the relevant Nanobodies, unless explicitly indicated otherwise.
  • polypeptides of the invention contain two or more Nanobodies and one or more further amino acid sequences (as mentioned herein).
  • Nanobodies may for example be Nanobodies that are directed against a serum protein, and in particular a human serum protein, such as human serum albumin, thyroxine-binding protein, (human) transferrin, fibrinogen, an immunoglobulin such as IgG, IgE or IgM, or against one of the serum proteins listed in WO 04/003019.
  • a human serum protein such as human serum albumin, thyroxine-binding protein, (human) transferrin, fibrinogen, an immunoglobulin such as IgG, IgE or IgM, or against one of the serum proteins listed in WO 04/003019.
  • Nanobodies that can bind to serum albumin (and in particular human serum albumin) or to IgG (and in particular human IgG, see for example Nanobody VH-I described in the review by Muyldermans, supra) are particularly preferred (although for example, for experiments in mice or primates, Nanobodies against or cross-reactive with mouse serum albumin (MSA) or serum albumin from said primate, respectively, can be used. However, for pharmaceutical use, Nanobodies against human serum albumin or human IgG will usually be preferred).
  • Nanobodies that provide for increased half-life and that can be used in the polypeptides of the invention include the Nanobodies directed against serum albumin that are described in WO 04/041865, in WO 06/122787 and in the further patent applications by Ablynx N.V., such as those mentioned above.
  • the some preferred Nanobodies that provide for increased half-life for use in the present invention include Nanobodies that can bind to amino acid residues on (human) serum albumin that are not involved in binding of serum albumin to FcRn (see for example WO 06/0122787); Nanobodies that are capable of binding to amino acid residues on serum albumin that do not form part of domain III of serum albumin (see for example WO 06/0122787); Nanobodies that have or can provide an increased half-life (see for example the US provisional application 60/843,349 by Ab lynx N.
  • Nanobodies against human serum albumin that are cross-reactive with serum albumin from at least one species of mammal, and in particular with at least one species of primate such as, without limitation, monkeys from the genus Macaca (such as, and in particular, cynomolgus monkeys ⁇ Macaca fascicularis) and/or rhesus monkeys ⁇ Macaca mulatto)) and baboon ⁇ Papio ursinus)
  • primate such as, without limitation, monkeys from the genus Macaca (such as, and in particular, cynomolgus monkeys ⁇ Macaca fascicularis) and/or rhesus monkeys ⁇ Macaca mulatto)
  • primate such as, without limitation, monkeys from the genus Macaca (such as, and in particular, cynomolgus monkeys ⁇ Macaca fascicularis) and/or rhesus monkeys ⁇ Macaca mulatto)
  • Nanobodies that can bind to serum albumin in a pH independent manner see for example the US provisional application 60/850,774 by Ablynx N.V. mentioned herein
  • Nanobodies that are conditional binders see for example the US provisional application 60/850,775 by Ablynx N.V.; see also PCT/EP2007/060850.
  • the polypeptides of the invention contain, besides the one or more Nanobodies of the invention, at least one Nanobody against human serum albumin.
  • any polypeptides of the invention with increased half-life that contain one or more Nanobodies of the invention, and any derivatives of Nanobodies of the invention or of such polypeptides that have an increased half-life preferably have a half-life that is at least 1.5 times, preferably at least 2 times, such as at least 5 times, for example at least 10 times or more than 20 times, greater than the half-life of the corresponding Nanobody of the invention per se.
  • such a derivative or polypeptides with increased half-life may have a half-life that is increased with more than 1 hours, preferably more than 2 hours, more preferably more than 6 hours, such as more than 12 hours, or even more than 24, 48 or 72 hours, compared to the corresponding Nanobody of the invention per se.
  • such derivatives or polypeptides may exhibit a serum half-life in human of at least about 12 hours, preferably at least 24 hours, more preferably at least 48 hours, even more preferably at least 72 hours or more.
  • such derivatives or polypeptides may have a half-life of at least 5 days (such as about 5 to 10 days), preferably at least 9 days (such as about 9 to 14 days), more preferably at least about 10 days (such as about 10 to 15 days), or at least about 11 days (such as about 11 to 16 days), more preferably at least about 12 days (such as about 12 to 18 days or more), or more than 14 days (such as about 14 to 19 days).
  • polypeptides are capable of binding to one or more molecules which can increase the half-life of the polypeptide in vivo.
  • polypeptides of the invention are stabilised in vivo and their half-life increased by binding to molecules which resist degradation and/or clearance or sequestration. Typically, such molecules are naturally occurring proteins which themselves have a long half-life in vivo.
  • a multispecific polypeptide of the invention comprises at least one Nanobody of the invention and at least one Nanobody that directs the polypeptide of the invention towards, and/or that allows the polypeptide of the invention to penetrate or to enter into specific organs, tissues, cells, or parts or compartments of cells, and/or that allows the Nanobody to penetrate or cross a biological barrier such as a cell membrane, a cell layer such as a layer of epithelial cells, a tumor including solid tumors, or the blood-brain-barrier.
  • Nanobodies examples include Nanobodies that are directed towards specific cell-surface proteins, markers or epitopes of the desired organ, tissue or cell (for example cell-surface markers associated with tumor cells), and the single- domain brain targeting antibody fragments described in WO 02/057445 and WO 06/040153, of which FC44 (SEQ ID NO: 189 of WO 06/040153) and FC5 (SEQ ID NO: 190 of WO 06/040154) are preferred examples.
  • the one or more Nanobodies and the one or more polypeptides may be directly linked to each other (as for example described in WO 99/23221) and/or may be linked to each other via one or more suitable spacers or linkers, or any combination thereof.
  • Suitable spacers or linkers for use in multivalent and multispecific polypeptides will be clear to the skilled person, and may generally be any linker or spacer used in the art to link amino acid sequences.
  • said linker or spacer is suitable for use in constructing proteins or polypeptides that are intended for pharmaceutical use.
  • Some particularly preferred spacers include the spacers and linkers that are used in the art to link antibody fragments or antibody domains.
  • linkers mentioned in the general background art cited above include the linkers mentioned in the general background art cited above, as well as for example linkers that are used in the art to construct diabodies or ScFv fragments (in this respect, however, its should be noted that, whereas in diabodies and in ScFv fragments, the linker sequence used should have a length, a degree of flexibility and other properties that allow the pertinent VH and VL domains to come together to form the complete antigen-binding site, there is no particular limitation on the length or the flexibility of the linker used in the polypeptide of the invention, since each Nanobody by itself forms a complete antigen-binding site).
  • a linker may be a suitable amino acid sequence, and in particular amino acid sequences of between 1 and 50, preferably between 1 and 30, such as between 1 and 10 amino acid residues.
  • amino acid sequences include gly-ser linkers, for example of the type (gly x ser y ) z , such as (for example (gly 4 ser)3 or (gly3ser 2 )3, as described in WO 99/42077 and the GS30, GS15, GS9 and GS7 linkers described in the applications by Ablynx mentioned herein (see for example WO 06/040153 and WO
  • hinge-like regions such as the hinge regions of naturally occurring heavy chain antibodies or similar sequences (such as described in WO 94/04678 ).
  • linkers are poly-alanine (such as AAA), as well as the linkers GS30 (SEQ ID NO: 85 in WO 06/122825) and GS9 (SEQ ID NO: 84 in WO 06/122825).
  • linkers generally comprise organic compounds or polymers, in particular those suitable for use in proteins for pharmaceutical use.
  • poly(ethyleneglycol) moieties have been used to link antibody domains, see for example WO 04/081026.
  • the length, the degree of flexibility and/or other properties of the linker(s) used may have some influence on the properties of the final polypeptide of the invention, including but not limited to the affinity, specificity or avidity for a metalloproteinase from the ADAM family, or for one or more of the other antigens.
  • the skilled person will be able to determine the optimal linker(s) for use in a specific polypeptide of the invention, optionally after some limited routine experiments.
  • the length and flexibility of the linker are preferably such that it allows each Nanobody of the invention present in the polypeptide to bind to the antigenic determinant on each of the subunits of the multimer.
  • the length and flexibility of the linker are preferably such that it allows each Nanobody to bind to its intended antigenic determinant.
  • linker(s) used confer one or more other favourable properties or functionality to the polypeptides of the invention, and/or provide one or more sites for the formation of derivatives and/or for the attachment of functional groups (e.g. as described herein for the derivatives of the Nanobodies of the invention).
  • linkers containing one or more charged amino acid residues can provide improved hydrophilic properties
  • linkers that form or contain small epitopes or tags can be used for the purposes of detection, identification and/or purification.
  • linkers when two or more linkers are used in the polypeptides of the invention, these linkers may be the same or different. Again, based on the disclosure herein, the skilled person will be able to determine the optimal linkers for use in a specific polypeptide of the invention, optionally after some limited routine experiments.
  • a polypeptide of the invention will be a linear polypeptide.
  • the invention in its broadest sense is not limited thereto.
  • a linker with three or more "arms", which each "arm” being linked to a Nanobody, so as to provide a "star-shaped" construct. It is also possible, although usually less preferred, to use circular constructs.
  • the invention also comprises derivatives of the polypeptides of the invention, which may be essentially analogous to the derivatives of the Nanobodies of the invention, i.e. as described herein.
  • the invention also comprises proteins or polypeptides that "essentially consist” of a polypeptide of the invention (in which the wording "essentially consist of has essentially the same meaning as indicated hereinabove).
  • the polypeptide of the invention is in essentially isolated from, as defined herein.
  • the amino acid sequences, Nanobodies, polypeptides and nucleic acids of the invention can be prepared in a manner known per se, as will be clear to the skilled person from the further description herein.
  • the Nanobodies and polypeptides of the invention can be prepared in any manner known per se for the preparation of antibodies and in particular for the preparation of antibody fragments (including but not limited to (single) domain antibodies and ScFv fragments).
  • Some preferred, but non-limiting methods for preparing the amino acid sequences, Nanobodies, polypeptides and nucleic acids include the methods and techniques described herein.
  • one particularly useful method for preparing an amino acid sequence, Nanobody and/or a polypeptide of the invention generally comprises the steps of: i) the expression, in a suitable host cell or host organism (also referred to herein as a "host of the invention") or in another suitable expression system of a nucleic acid that encodes said amino acid sequence, Nanobody or polypeptide of the invention (also referred to herein as a "nucleic acid of the invention”), optionally followed by: ii) isolating and/or purifying the amino acid sequence, Nanobody or polypeptide of the invention thus obtained.
  • such a method may comprise the steps of: i) cultivating and/or maintaining a host of the invention under conditions that are such that said host of the invention expresses and/or produces at least one amino acid sequence, Nanobody and/or polypeptide of the invention; optionally followed by: ii) isolating and/or purifying the amino acid sequence, Nanobody or polypeptide of the invention thus obtained.
  • a nucleic acid of the invention can be in the form of single or double stranded DNA or RNA, and is preferably in the form of double stranded DNA.
  • the nucleotide sequences of the invention may be genomic DNA, cDNA or synthetic DNA (such as DNA with a codon usage that has been specifically adapted for expression in the intended host cell or host organism).
  • the nucleic acid of the invention is in essentially isolated from, as defined herein.
  • the nucleic acid of the invention may also be in the form of, be present in and/or be part of a vector, such as for example a plasmid, cosmid or YAC, which again may be in essentially isolated form.
  • nucleic acids of the invention can be prepared or obtained in a manner known per se, based on the information on the amino acid sequences for the polypeptides of the invention given herein, and/or can be isolated from a suitable natural source.
  • nucleotide sequences encoding naturally occurring VHH domains can for example be subjected to site-directed mutagenesis, so at to provide a nucleic acid of the invention encoding said analog.
  • nucleic acid of the invention also several nucleotide sequences, such as at least one nucleotide sequence encoding a Nanobody and for example nucleic acids encoding one or more linkers can be linked together in a suitable manner.
  • nucleic acids of the invention may for instance include, but are not limited to, automated DNA synthesis; site- directed mutagenesis; combining two or more naturally occurring and/or synthetic sequences (or two or more parts thereof), introduction of mutations that lead to the expression of a truncated expression product; introduction of one or more restriction sites (e.g. to create cassettes and/or regions that may easily be digested and/or ligated using suitable restriction enzymes), and/or the introduction of mutations by means of a PCR reaction using one or more "mismatched" primers, using for example a sequence of a naturally occurring form of a metalloproteinase from the ADAM family as a template.
  • the nucleic acid of the invention may also be in the form of, be present in and/or be part of a genetic construct, as will be clear to the person skilled in the art.
  • Such genetic constructs generally comprise at least one nucleic acid of the invention that is optionally linked to one or more elements of genetic constructs known per se, such as for example one or more suitable regulatory elements (such as a suitable promoter(s), enhancer(s), terminator(s), etc.) and the further elements of genetic constructs referred to herein.
  • suitable regulatory elements such as a suitable promoter(s), enhancer(s), terminator(s), etc.
  • Such genetic constructs comprising at least one nucleic acid of the invention will also be referred to herein as "genetic constructs of the invention”.
  • the genetic constructs of the invention may be DNA or RNA, and are preferably double-stranded DNA.
  • the genetic constructs of the invention may also be in a form suitable for transformation of the intended host cell or host organism, in a form suitable for integration into the genomic DNA of the intended host cell or in a form suitable for independent replication, maintenance and/or inheritance in the intended host organism.
  • the genetic constructs of the invention may be in the form of a vector, such as for example a plasmid, cosmid, YAC, a viral vector or transposon.
  • the vector may be an expression vector, i.e. a vector that can provide for expression in vitro and/or in vivo (e.g. in a suitable host cell, host organism and/or expression system).
  • a genetic construct of the invention comprises i) at least one nucleic acid of the invention; operably connected to ii) one or more regulatory elements, such as a promoter and optionally a suitable terminator; and optionally also iii) one or more further elements of genetic constructs known per se; in which the terms "regulatory element”, “promoter”, “terminator” and “operably connected” have their usual meaning in the art (as further described herein); and in which said "further elements” present in the genetic constructs may for example be 3'- or 5'-UTR sequences, leader sequences, selection markers, expression markers/reporter genes, and/or elements that may facilitate or increase (the efficiency of) transformation or integration.
  • nucleotide sequences of the invention of interest are to be expressed (e.g. via constitutive, transient or inducible expression); and/or the transformation technique to be used.
  • regulatory sequences, promoters and terminators known per se for the expression and production of antibodies and antibody fragments may be used in an essentially analogous manner.
  • said at least one nucleic acid of the invention and said regulatory elements, and optionally said one or more further elements are "operably linked" to each other, by which is generally meant that they are in a functional relationship with each other.
  • a promoter is considered “operably linked” to a coding sequence if said promoter is able to initiate or otherwise control/regulate the transcription and/or the expression of a coding sequence (in which said coding sequence should be understood as being "under the control of' said promoter).
  • the regulatory and further elements of the genetic constructs of the invention are such that they are capable of providing their intended biological function in the intended host cell or host organism.
  • a promoter, enhancer or terminator should be "operable" in the intended host cell or host organism, by which is meant that (for example) said promoter should be capable of initiating or otherwise controlling/regulating the transcription and/or the expression of a nucleotide sequence - e.g. a coding sequence - to which it is operably linked (as defined herein).
  • promoters include, but are not limited to, promoters known per se for the expression in the host cells mentioned herein; and in particular promoters for the expression in the bacterial cells, such as those mentioned herein and/or those used in the Examples.
  • a selection marker should be such that it allows - i.e. under appropriate selection conditions - host cells and/or host organisms that have been (successfully) transformed with the nucleotide sequence of the invention to be distinguished from host cells/organisms that have not been (successfully) transformed.
  • Some preferred, but non -limiting examples of such markers are genes that provide resistance against antibiotics (such as kanamycin or ampicillin), genes that provide for temperature resistance, or genes that allow the host cell or host organism to be maintained in the absence of certain factors, compounds and/or (food) components in the medium that are essential for survival of the non-transformed cells or organisms.
  • leader sequence should be such that - in the intended host cell or host organism - it allows for the desired post-translational modifications and/or such that it directs the transcribed mRNA to a desired part or organelle of a cell.
  • a leader sequence may also allow for secretion of the expression product from said cell.
  • the leader sequence may be any pro-, pre-, or prepro -sequence operable in the host cell or host organism.
  • Leader sequences may not be required for expression in a bacterial cell.
  • leader sequences known per se for the expression and production of antibodies and antibody fragments may be used in an essentially analogous manner.
  • An expression marker or reporter gene should be such that - in the host cell or host organism - it allows for detection of the expression of (a gene or nucleotide sequence present on) the genetic construct.
  • An expression marker may optionally also allow for the localisation of the expressed product, e.g. in a specific part or organelle of a cell and/or in (a) specific cell(s), tissue(s), organ(s) or part(s) of a multicellular organism.
  • Such reporter genes may also be expressed as a protein fusion with the amino acid sequence of the invention. Some preferred, but non-limiting examples include fluorescent proteins such as GFP.
  • suitable promoters, terminator and further elements include those that can be used for the expression in the host cells mentioned herein; and in particular those that are suitable for expression in bacterial cells, such as those mentioned herein and/or those used in the Examples below.
  • suitable promoters, selection markers, leader sequences, expression markers and further elements that may be present/used in the genetic constructs of the invention - such as terminators, transcriptional and/or translational enhancers and/or integration factors - reference is made to the general handbooks such as Sambrook et al. and Ausubel et al.
  • the genetic constructs of the invention may generally be provided by suitably linking the nucleotide sequence(s) of the invention to the one or more further elements described above, for example using the techniques described in the general handbooks such as Sambrook et al. and Ausubel et al., mentioned above. Often, the genetic constructs of the invention will be obtained by inserting a nucleotide sequence of the invention in a suitable (expression) vector known per se. Some preferred, but non-limiting examples of suitable expression vectors are those used in the Examples below, as well as those mentioned herein.
  • the nucleic acids of the invention and/or the genetic constructs of the invention may be used to transform a host cell or host organism, i.e. for expression and/or production of the amino acid sequence, Nanobody or polypeptide of the invention.
  • Suitable hosts or host cells will be clear to the skilled person, and may for example be any suitable fungal, prokaryotic or eukaryotic cell or cell line or any suitable fungal, prokaryotic or eukaryotic organism, for example: a bacterial strain, including but not limited to gram-negative strains such as strains of Escherichia coli; of Proteus, for example of Proteus mirabilis; of Pseudomonas, for example of Pseudomonas fluorescens; and gram-positive strains such as strains of Bacillus, for example of Bacillus subtilis or of Bacillus brevis; ofStreptomyces, for example of Streptomyces lividans; of Staphylococcus
  • Nanobodies and polypeptides of the invention can also be introduced and expressed in one or more cells, tissues or organs of a multicellular organism, for example for prophylactic and/or therapeutic purposes (e.g.
  • nucleotide sequences of the invention may be introduced into the cells or tissues in any suitable way, for example as such (e.g. using liposomes) or after they have been inserted into a suitable gene therapy vector (for example derived from retroviruses such as adenovirus, or parvoviruses such as adeno-associated virus).
  • a suitable gene therapy vector for example derived from retroviruses such as adenovirus, or parvoviruses such as adeno-associated virus.
  • such gene therapy may be performed in vivo and/or in situ in the body of a patient by administering a nucleic acid of the invention or a suitable gene therapy vector encoding the same to the patient or to specific cells or a specific tissue or organ of the patient; or suitable cells (often taken from the body of the patient to be treated, such as explanted lymphocytes, bone marrow aspirates or tissue biopsies) may be treated in vitro with a nucleotide sequence of the invention and then be suitably (re-)introduced into the body of the patient. All this can be performed using gene therapy vectors, techniques and delivery systems which are well known to the skilled person, and for example described in Culver, K.
  • Nanobodies for expression of the Nanobodies in a cell, they may also be expressed as so-called “intrabodies”, as for example described in WO 94/02610, WO 95/22618 and US-A-7004940; WO 03/014960; in Cattaneo, A. & Biocca, S. (1997) Intracellular Antibodies: Development and Applications. Austin and Springer- Verlag; and in Kontermann, Methods 34, (2004), 163-170.
  • intraabodies as for example described in WO 94/02610, WO 95/22618 and US-A-7004940; WO 03/014960; in Cattaneo, A. & Biocca, S. (1997) Intracellular Antibodies: Development and Applications. Austin and Springer- Verlag; and in Kontermann, Methods 34, (2004), 163-170.
  • Nanobodies and polypeptides of the invention can for example also be produced in the milk of transgenic mammals, for example in the milk of rabbits, cows, goats or sheep (see for example US-A-6,741,957, US-A-6,304,489 and US-A- 6,849,992 for general techniques for introducing transgenes into mammals), in plants or parts of plants including but not limited to their leaves, flowers, fruits, seed, roots or tubers (for example in tobacco, maize, soybean or alfalfa) or in for example pupae of the silkworm Bombix mori.
  • amino acid sequences, Nanobodies and polypeptides of the invention can also be expressed and/or produced in cell-free expression systems, and suitable examples of such systems will be clear to the skilled person.
  • suitable examples include expression in the wheat germ system; in rabbit reticulocyte lysates; or in the E. coli Zubay system.
  • Nanobodies As mentioned above, one of the advantages of the use of Nanobodies is that the polypeptides based thereon can be prepared through expression in a suitable bacterial system, and suitable bacterial expression systems, vectors, host cells, regulatory elements, etc., will be clear to the skilled person, for example from the references cited above. It should however be noted that the invention in its broadest sense is not limited to expression in bacterial systems.
  • an (in vivo or in vitro) expression system such as a bacterial expression system, is used that provides the polypeptides of the invention in a form that is suitable for pharmaceutical use, and such expression systems will again be clear to the skilled person.
  • polypeptides of the invention suitable for pharmaceutical use can be prepared using techniques for peptide synthesis.
  • preferred heterologous hosts for the (industrial) production of Nanobodies or Nanobody-containing protein therapeutics include strains ofii. coli, Pichiapastoris, S. cerevisiae that are suitable for large scale expression/production/fermentation, and in particular for large scale pharmaceutical (i.e. GMP grade) expression/production/fermentation. Suitable examples of such strains will be clear to the skilled person.
  • Such strains and production/expression systems are also made available by companies such as Biovitrum (Uppsala, Sweden).
  • mammalian cell lines in particular Chinese hamster ovary (CHO) cells, can be used for large scale expression/production/fermentation, and in particular for large scale pharmaceutical expression/production/fermentation.
  • CHO Chinese hamster ovary
  • the choice of the specific expression system would depend in part on the requirement for certain post-translational modifications, more specifically glycosylation.
  • the production of a Nanobody-containing recombinant protein for which glycosylation is desired or required would necessitate the use of mammalian expression hosts that have the ability to glycosylate the expressed protein.
  • the glycosylation pattern obtained i.e. the kind, number and position of residues attached
  • the cell or cell line is used for the expression.
  • a human cell or cell line is used (i.e.
  • the amino acid sequence, Nanobody or polypeptide of the invention is glycosylated. According to another non-limiting aspect of the invention, the amino acid sequence, Nanobody or polypeptide of the invention is non-glycosylated.
  • the amino acid sequence, Nanobody or polypeptide of the invention is produced in a bacterial cell, in particular a bacterial cell suitable for large scale pharmaceutical production, such as cells of the strains mentioned above.
  • the amino acid sequence, Nanobody or polypeptide of the invention is produced in a yeast cell, in particular a yeast cell suitable for large scale pharmaceutical production, such as cells of the species mentioned above.
  • the amino acid sequence, Nanobody or polypeptide of the invention is produced in a mammalian cell, in particular in a human cell or in a cell of a human cell line, and more in particular in a human cell or in a cell of a human cell line that is suitable for large scale pharmaceutical production, such as the cell lines mentioned hereinabove.
  • Nanobodies and the polypeptides of the invention can be produced either intracellullarly (e.g. in the cytosol, in the periplasma or in inclusion bodies) and then isolated from the host cells and optionally further purified; or can be produced extracellularly (e.g. in the medium in which the host cells are cultured) and then isolated from the culture medium and optionally further purified.
  • extracellular production is usually preferred since this considerably facilitates the further isolation and downstream processing of the Nanobodies and proteins obtained.
  • Bacterial cells such as the strains of E.
  • coli normally do not secrete proteins extracellularly, except for a few classes of proteins such as toxins and hemolysin, and secretory production inii. coli refers to the translocation of proteins across the inner membrane to the periplasmic space.
  • Periplasmic production provides several advantages over cytosolic production. For example, the N-terminal amino acid sequence of the secreted product can be identical to the natural gene product after cleavage of the secretion signal sequence by a specific signal peptidase. Also, there appears to be much less protease activity in the periplasm than in the cytoplasm. In addition, protein purification is simpler due to fewer contaminating proteins in the periplasm.
  • Another advantage is that correct disulfide bonds may form because the periplasm provides a more oxidative environment than the cytoplasm. Proteins overexpressed in E. coli are often found in insoluble aggregates, so-called inclusion bodies. These inclusion bodies may be located in the cytosol or in the periplasm; the recovery of biologically active proteins from these inclusion bodies requires a denaturation/refolding process. Many recombinant proteins, including therapeutic proteins, are recovered from inclusion bodies. Alternatively, as will be clear to the skilled person, recombinant strains of bacteria that have been genetically modified so as to secrete a desired protein, and in particular an amino acid sequence, Nanobody or a polypeptide of the invention, can be used.
  • the amino acid sequence, Nanobody or polypeptide of the invention is an amino acid sequence, Nanobody or polypeptide that has been produced intracellularly and that has been isolated from the host cell, and in particular from a bacterial cell or from an inclusion body in a bacterial cell.
  • the amino acid sequence, Nanobody or polypeptide of the invention is an amino acid sequence, Nanobody or polypeptide that has been produced extracellularly, and that has been isolated from the medium in which the host cell is cultivated.
  • Some preferred, but non-limiting promoters for use with these host cells include, for expression in E.
  • lac promoter and derivatives thereof such as the lacUV5 promoter); arabinose promoter; left- (PL) and rightward (PR) promoter of phage lambda; promoter of the trp operon; hybrid lac/trp promoters (tac and trc); T7-promoter (more specifically that of T7-phage gene 10) and other T-phage promoters; promoter of the TnIO tetracycline resistance gene; engineered variants of the above promoters that include one or more copies of an extraneous regulatory operator sequence; for expression in S.
  • ADHl alcohol dehydrogenase 1
  • ENO enolase
  • CYCl cytochrome c iso-1
  • GAPDH glycosyl-3 -phosphate dehydrogenase
  • PGKl phosphoglycerate kinase
  • PYKl pyruvate kinase
  • GAL 1,10,7 galactose metabolic enzymes
  • ADH2 alcohol dehydrogenase 2
  • PHO5 acid phosphatase
  • CUPl copper metallothionein
  • heterologous CaMV (cauliflower mosaic virus 35 S promoter); for expression in Pichia pastoris: the AOXl promoter (alcohol oxidase I); - for expression in mammalian cells: human cytomegalovirus (hCMV) immediate early enhancer/promoter; human cytomegalovirus (hCMV) immediate early promoter variant that contains two tetracycline operator sequences such that the promoter can be regulated by the Tet repressor; Herpes Simplex Virus thymidine kinase (TK) promoter; Rous Sarcoma Virus long terminal repeat (RSV LTR) enhancer/promoter; elongation factor l ⁇ (hEF-l ⁇ ) promoter from human, chimpanzee, mouse or
  • vectors for use with these host cells include: vectors for expression in mammalian cells: pMAMneo (Clontech), pcDNA3
  • Some preferred, but non-limiting secretory sequences for use with these host cells include: for use in bacterial cells such as E. coli: PeIB, BIa, OmpA, OmpC, OmpF, OmpT, StII, PhoA, PhoE, MaIE, Lpp, LamB, and the like; TAT signal peptide, hemolysin C- terminal secretion signal; for use in yeast: ⁇ -mating factor prepro-sequence, phosphatase (phol), invertase (Sue), etc.; for use in mammalian cells: indigenous signal in case the target protein is of eukaryotic origin; murine Ig ⁇ -chain V-J2-C signal peptide; etc.
  • Suitable techniques for transforming a host or host cell of the invention will be clear to the skilled person and may depend on the intended host cell/host organism and the genetic construct to be used. Reference is again made to the handbooks and patent applications mentioned above.
  • a step for detecting and selecting those host cells or host organisms that have been successfully transformed with the nucleotide sequence/genetic construct of the invention may be performed. This may for instance be a selection step based on a selectable marker present in the genetic construct of the invention or a step involving the detection of the amino acid sequence of the invention, e.g. using specific antibodies.
  • the transformed host cell (which may be in the form or a stable cell line) or host organisms (which may be in the form of a stable mutant line or strain) form further aspects of the present invention.
  • these host cells or host organisms are such that they express, or are (at least) capable of expressing (e.g. under suitable conditions), an amino acid sequence, Nanobody or polypeptide of the invention (and in case of a host organism: in at least one cell, part, tissue or organ thereof).
  • the invention also includes further generations, progeny and/or offspring of the host cell or host organism of the invention, that may for instance be obtained by cell division or by sexual or asexual reproduction.
  • the transformed host cell or transformed host organism may generally be kept, maintained and/or cultured under conditions such that the (desired) amino acid sequence, Nanobody or polypeptide of the invention is expressed/produced. Suitable conditions will be clear to the skilled person and will usually depend upon the host cell/host organism used, as well as on the regulatory elements that control the expression of the (relevant) nucleotide sequence of the invention. Again, reference is made to the handbooks and patent applications mentioned above in the paragraphs on the genetic constructs of the invention.
  • suitable conditions may include the use of a suitable medium, the presence of a suitable source of food and/or suitable nutrients, the use of a suitable temperature, and optionally the presence of a suitable inducing factor or compound (e.g. when the nucleotide sequences of the invention are under the control of an inducible promoter); all of which may be selected by the skilled person.
  • a suitable inducing factor or compound e.g. when the nucleotide sequences of the invention are under the control of an inducible promoter
  • the amino acid sequences of the invention may be expressed in a constitutive manner, in a transient manner, or only when suitably induced.
  • amino acid sequence, Nanobody or polypeptide of the invention may (first) be generated in an immature form (as mentioned above), which may then be subjected to post-translational modification, depending on the host cell/host organism used.
  • amino acid sequence, Nanobody or polypeptide of the invention may be glycosylated, again depending on the host cell/host organism used.
  • the amino acid sequence, Nanobody or polypeptide of the invention may then be isolated from the host cell/host organism and/or from the medium in which said host cell or host organism was cultivated, using protein isolation and/or purification techniques known per se, such as (preparative) chromatography and/or electrophoresis techniques, differential precipitation techniques, affinity techniques (e.g. using a specific, cleavable amino acid sequence fused with the amino acid sequence, Nanobody or polypeptide of the invention) and/or preparative immunological techniques (i.e. using antibodies against the amino acid sequence to be isolated).
  • protein isolation and/or purification techniques known per se such as (preparative) chromatography and/or electrophoresis techniques, differential precipitation techniques, affinity techniques (e.g. using a specific, cleavable amino acid sequence fused with the amino acid sequence, Nanobody or polypeptide of the invention) and/or preparative immunological techniques (i.e. using antibodies against the amino acid sequence to be isolated).
  • the polypeptides of the invention may be formulated as a pharmaceutical preparation or compositions comprising at least one polypeptide of the invention and at least one pharmaceutically acceptable carrier, diluent or excipient and/or adjuvant, and optionally one or more further pharmaceutically active polypeptides and/or compounds.
  • a formulation may be in a form suitable for oral administration, for parenteral administration (such as by intravenous, intramuscular or subcutaneous injection or intravenous infusion), for topical administration, for administration by inhalation, by a skin patch, by an implant, by a suppository, etc.
  • the invention relates to a pharmaceutical composition that contains at least one amino acid of the invention, at least one Nanobody of the invention or at least one polypeptide of the invention and at least one suitable carrier, diluent or excipient (i.e. suitable for pharmaceutical use), and optionally one or more further active substances.
  • the amino acid sequences, Nanobodies and polypeptides of the invention can be formulated and administered in any suitable manner known per se, for which reference is for example made to the general background art cited above (and in particular to WO 04/041862, WO 04/041863, WO 04/041865 and WO 04/041867) as well as to the standard handbooks, such as Remington's Pharmaceutical Sciences, 18 th Ed., Mack Publishing Company, USA (1990) or Remington, the Science and Practice of Pharmacy, 21st Edition, Lippincott Williams and Wilkins (2005).
  • amino acid sequences, Nanobodies and polypeptides of the invention may be formulated and administered in any manner known per se for conventional antibodies and antibody fragments (including ScFv's and diabodies) and other pharmaceutically active proteins.
  • Such formulations and methods for preparing the same will be clear to the skilled person, and for example include preparations suitable for parenteral administration (for example intravenous, intraperitoneal, subcutaneous, intramuscular, intraluminal, intra-arterial or intrathecal administration) or for topical (i.e. transdermal or intradermal) administration.
  • Preparations for parenteral administration may for example be sterile solutions, suspensions, dispersions or emulsions that are suitable for infusion or injection.
  • Suitable carriers or diluents for such preparations for example include, without limitation, sterile water and aqueous buffers and solutions such as physiological phosphate-buffered saline, Ringer's solutions, dextrose solution, and Hank's solution; water oils; glycerol; ethanol; glycols such as propylene glycol or as well as mineral oils, animal oils and vegetable oils, for example peanut oil, soybean oil, as well as suitable mixtures thereof.
  • aqueous solutions or suspensions will be preferred.
  • the amino acid sequences, Nanobodies and polypeptides of the invention can also be administered using gene therapy methods of delivery. See, e.g., U.S. Patent No. 5,399,346, which is incorporated by reference in its entirety.
  • gene therapy methods of delivery primary cells transfected with the gene encoding an amino acid sequence, Nanobody or polypeptide of the invention can additionally be transfected with tissue specific promoters to target specific organs, tissue, grafts, tumors, or cells and can additionally be transfected with signal and stabilization sequences for subcellularly localized expression.
  • amino acid sequences, Nanobodies and polypeptides of the invention may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet.
  • a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier.
  • the amino acid sequences, Nanobodies and polypeptides of the invention may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • compositions and preparations should contain at least 0.1% of the amino acid sequence, Nanobody or polypeptide of the invention. Their percentage in the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form. The amount of the amino acid sequence, Nanobody or polypeptide of the invention in such therapeutically useful compositions is such that an effective dosage level will be obtained.
  • the tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added.
  • a liquid carrier such as a vegetable oil or a polyethylene glycol.
  • any material may be present as coatings or to otherwise modify the physical form of the solid unit dosage form.
  • tablets, pills, or capsules may be coated with gelatin, wax, shellac or sugar and the like.
  • a syrup or elixir may contain the amino acid sequences, Nanobodies and polypeptides of the invention, sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor.
  • any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed.
  • the amino acid sequences, Nanobodies and polypeptides of the invention may be incorporated into sustained-release preparations and devices.
  • Preparations and formulations for oral administration may also be provided with an enteric coating that will allow the constructs of the invention to resist the gastric environment and pass into the intestines. More generally, preparations and formulations for oral administration may be suitably formulated for delivery into any desired part of the gastrointestinal tract. In addition, suitable suppositories may be used for delivery into the gastrointestinal tract.
  • the amino acid sequences, Nanobodies and polypeptides of the invention may also be administered intravenously or intraperitoneally by infusion or injection.
  • Solutions of the amino acid sequences, Nanobodies and polypeptides of the invention or their salts can be prepared in water, optionally mixed with a nontoxic surfactant.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes.
  • the liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the amino acid sequences, Nanobodies and polypeptides of the invention in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization.
  • the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
  • the amino acid sequences, Nanobodies and polypeptides of the invention maybe applied in pure form, i.e., when they are liquids. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid.
  • Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like.
  • Useful liquid carriers include water, hydroxyalkyls or glycols or water-alcohol/glycol blends, in which the amino acid sequences, Nanobodies and polypeptides of the invention can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants.
  • Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use.
  • the resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.
  • Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
  • Examples of useful dermatological compositions which can be used to deliver the amino acid sequences, Nanobodies and polypeptides of the invention to the skin are known to the art; for example, see Jacquet et al. (U.S. Pat. No. 4,608,392), Geria (U.S. Pat. No. 4,992,478), Smith et al. (U.S. Pat. No. 4,559,157) and Wortzman (U.S. Pat. No. 4,820,508).
  • Useful dosages of the amino acid sequences, Nanobodies and polypeptides of the invention can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949.
  • the concentration of the amino acid sequences, Nanobodies and polypeptides of the invention in a liquid composition will be from about 0.1-25 wt-%, preferably from about 0.5-10 wt-%.
  • concentration in a semi-solid or solid composition such as a gel or a powder will be about 0.1-5 wt-%, preferably about 0.5-2.5 wt-
  • the amount of the amino acid sequences, Nanobodies and polypeptides of the invention required for use in treatment will vary not only with the particular amino acid sequence, Nanobody or polypeptide selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician. Also the dosage of the amino acid sequences, Nanobodies and polypeptides of the invention varies depending on the target cell, tumor, tissue, graft, or organ.
  • the desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
  • the sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
  • An administration regimen could include long-term, daily treatment.
  • long-term is meant at least two weeks and preferably, several weeks, months, or years of duration. Necessary modifications in this dosage range may be determined by one of ordinary skill in the art using only routine experimentation given the teachings herein. See Remington's Pharmaceutical Sciences (Martin, E. W., ed. 4), Mack Publishing Co., Easton, PA. The dosage can also be adjusted by the individual physician in the event of any complication.
  • the invention in another aspect, relates to a method for the prevention and/or treatment of at least one ADAM -related diseases and disorders, said method comprising administering, to a subject in need thereof, a pharmaceutically active amount of an amino acid sequence of the invention, of aNanobody of the invention, of a polypeptide of the invention, and/or of a pharmaceutical composition comprising the same.
  • prevention and/or treatment not only comprises preventing and/or treating the disease, but also generally comprises preventing the onset of the disease, slowing or reversing the progress of disease, preventing or slowing the onset of one or more symptoms associated with the disease, reducing and/or alleviating one or more symptoms associated with the disease, reducing the severity and/or the duration of the disease and/or of any symptoms associated therewith and/or preventing a further increase in the severity of the disease and/or of any symptoms associated therewith, preventing, reducing or reversing any physiological damage caused by the disease, and generally any pharmacological action that is beneficial to the patient being treated.
  • the subject to be treated may be any warm-blooded animal, but is in particular a mammal, and more in particular a human being.
  • the subject to be treated will in particular be a person suffering from, or at risk of, the diseases and disorders mentioned herein.
  • the invention relates to a method for the prevention and/or treatment of at least one disease or disorder that is associated with a metalloproteinase from the ADAM family, with its biological or pharmacological activity, and/or with the biological pathways or signalling in which a metalloproteinase from the ADAM family is involved, said method comprising administering, to a subject in need thereof, a pharmaceutically active amount of an amino acid sequence of the invention, of a Nanobody of the invention, of a polypeptide of the invention, and/or of a pharmaceutical composition comprising the same.
  • the invention relates to a method for the prevention and/or treatment of at least one disease or disorder that can be treated by modulating a metalloproteinase from the ADAM family, its biological or pharmacological activity, and/or the biological pathways or signalling in which a metalloproteinase from the ADAM family is involved, said method comprising administering, to a subject in need thereof, a pharmaceutically active amount of an amino acid sequence of the invention, of a Nanobody of the invention, of a polypeptide of the invention, and/or of a pharmaceutical composition comprising the same.
  • said pharmaceutically effective amount may be an amount that is sufficient to modulate a metalloproteinase from the ADAM family, its biological or pharmacological activity, and/or the biological pathways or signalling in which a metalloproteinase from the ADAM family is involved; and/or an amount that provides a level of the amino acid sequence of the invention, of a Nanobody of the invention, of a polypeptide of the invention in the circulation that is sufficient to modulate a metalloproteinase from the ADAM family, its biological or pharmacological activity, and/or the biological pathways or signalling in which a metalloproteinase from the ADAM family is involved.
  • the invention furthermore relates to a method for the prevention and/or treatment of at least one disease or disorder that can be prevented and/or treated by administering an amino acid sequence of the invention, a Nanobody of the invention or a polypeptide of the invention to a patient, said method comprising administering, to a subject in need thereof, a pharmaceutically active amount of an amino acid sequence of the invention, of a Nanobody of the invention, of a polypeptide of the invention, and/or of a pharmaceutical composition comprising the same.
  • the invention relates to a method for the prevention and/or treatment of at least one disease or disorder chosen from the group consisting of the diseases and disorders listed herein, said method comprising administering, to a subject in need thereof, a pharmaceutically active amount of an amino acid sequence of the invention, of a Nanobody of the invention, of a polypeptide of the invention, and/or of a pharmaceutical composition comprising the same.
  • the invention relates to a method for immunotherapy, and in particular for passive immunotherapy, which method comprises administering, to a subject suffering from or at risk of the diseases and disorders mentioned herein, a pharmaceutically active amount of an amino acid sequence of the invention, of a Nanobody of the invention, of a polypeptide of the invention, and/or of a pharmaceutical composition comprising the same.
  • a pharmaceutically active amount of an amino acid sequence of the invention of a Nanobody of the invention, of a polypeptide of the invention, and/or of a pharmaceutical composition comprising the same.
  • the amino acid sequences, Nanobodies and/or polypeptides of the invention and/or the compositions comprising the same can be administered in any suitable manner, depending on the specific pharmaceutical formulation or composition to be used.
  • amino acid sequences, Nanobodies and/or polypeptides of the invention and/or the compositions comprising the same can for example be administered orally, intraperitoneally (e.g. intravenously, subcutaneously, intramuscularly, or via any other route of administration that circumvents the gastrointestinal tract), intranasally, transdermally, topically, by means of a suppository, by inhalation, again depending on the specific pharmaceutical formulation or composition to be used.
  • the clinician will be able to select a suitable route of administration and a suitable pharmaceutical formulation or composition to be used in such administration, depending on the disease or disorder to be prevented or treated and other factors well known to the clinician.
  • the amino acid sequences, Nanobodies and/or polypeptides of the invention and/or the compositions comprising the same are administered according to a regime of treatment that is suitable for preventing and/or treating the disease or disorder to be prevented or treated.
  • the clinician will generally be able to determine a suitable treatment regimen, depending on factors such as the disease or disorder to be prevented or treated, the severity of the disease to be treated and/or the severity of the symptoms thereof, the specific amino acid sequence, Nanobody or polypeptide of the invention to be used, the specific route of administration and pharmaceutical formulation or composition to be used, the age, gender, weight, diet, general condition of the patient, and similar factors well known to the clinician.
  • the treatment regimen will comprise the administration of one or more amino acid sequences, Nanobodies and/or polypeptides of the invention, or of one or more compositions comprising the same, in one or more pharmaceutically effective amounts or doses.
  • the specific amount(s) or doses to administered can be determined by the clinician, again based on the factors cited above.
  • the potency of the specific amino acid sequence, Nanobody and polypeptide of the invention to be used, the specific route of administration and the specific pharmaceutical formulation or composition used, the amino acid sequences, Nanobodies and polypeptides of the invention will generally be administered in an amount between 1 gram and 0.01 microgram per kg body weight per day, preferably between 0.1 gram and 0.1 microgram per kg body weight per day, such as about 1, 10, 100 or 1000 microgram per kg body weight per day, either continuously (e.g. by infusion), as a single daily dose or as multiple divided doses during the day.
  • the clinician will generally be able to determine a suitable daily dose, depending on the factors mentioned herein. It will also be clear that in specific cases, the clinician may choose to deviate from these amounts, for example on the basis of the factors cited above and his expert judgment. Generally, some guidance on the amounts to be administered can be obtained from the amounts usually administered for comparable conventional antibodies or antibody fragments against the same target administered via essentially the same route, taking into account however differences in affinity/avidity, efficacy, biodistribution, half-life and similar factors well known to the skilled person.
  • Nanobody or polypeptide of the invention a single amino acid sequence, Nanobody or polypeptide of the invention will be used. It is however within the scope of the invention to use two or more amino acid sequences, Nanobodies and/or polypeptides of the invention in combination.
  • the Nanobodies, amino acid sequences and polypeptides of the invention may also be used in combination with one or more further pharmaceutically active compounds or principles, i.e. as a combined treatment regimen, which may or may not lead to a synergistic effect. Again, the clinician will be able to select such further compounds or principles, as well as a suitable combined treatment regimen, based on the factors cited above and his expert judgement.
  • amino acid sequences, Nanobodies and polypeptides of the invention may be used in combination with other pharmaceutically active compounds or principles that are or can be used for the prevention and/or treatment of the diseases and disorders cited herein, as a result of which a synergistic effect may or may not be obtained.
  • examples of such compounds and principles, as well as routes, methods and pharmaceutical formulations or compositions for administering them will be clear to the clinician.
  • two or more substances or principles When two or more substances or principles are to be used as part of a combined treatment regimen, they can be administered via the same route of administration or via different routes of administration, at essentially the same time or at different times (e.g. essentially simultaneously, consecutively, or according to an alternating regime).
  • the substances or principles When the substances or principles are to be administered simultaneously via the same route of administration, they may be administered as different pharmaceutical formulations or compositions or part of a combined pharmaceutical formulation or composition, as will be clear to the skilled person.
  • each of the substances or principles may be administered in the same amount and according to the same regimen as used when the compound or principle is used on its own, and such combined use may or may not lead to a synergistic effect.
  • the effectiveness of the treatment regimen used according to the invention may be determined and/or followed in any manner known per se for the disease or disorder involved, as will be clear to the clinician.
  • the clinician will also be able, where appropriate and on a case-by-case basis, to change or modify a particular treatment regimen, so as to achieve the desired therapeutic effect, to avoid, limit or reduce unwanted side-effects, and/or to achieve an appropriate balance between achieving the desired therapeutic effect on the one hand and avoiding, limiting or reducing undesired side effects on the other hand.
  • the treatment regimen will be followed until the desired therapeutic effect is achieved and/or for as long as the desired therapeutic effect is to be maintained. Again, this can be determined by the clinician.
  • the invention relates to the use of an amino acid sequence, Nanobody or polypeptide of the invention in the preparation of a pharmaceutical composition for prevention and/or treatment of at least one ADAM -related diseases and disorders; and/or for use in one or more of the methods of treatment mentioned herein.
  • the subject to be treated may be any warm-blooded animal, but is in particular a mammal, and more in particular a human being.
  • the subject to be treated will in particular be a person suffering from, or at risk of, the diseases and disorders mentioned herein.
  • the invention also relates to the use of an amino acid sequence, Nanobody or polypeptide of the invention in the preparation of a pharmaceutical composition for the prevention and/or treatment of at least one disease or disorder that can be prevented and/or treated by administering an amino acid sequence, Nanobody or polypeptide of the invention to a patient.
  • the invention relates to the use of an amino acid sequence, Nanobody or polypeptide of the invention in the preparation of a pharmaceutical composition for the prevention and/or treatment of ADAM-related diseases and disorders, and in particular for the prevention and treatment of one or more of the diseases and disorders listed herein.
  • the one or more amino acid sequences, Nanobodies or polypeptides of the invention may also be suitably combined with one or more other active principles, such as those mentioned herein.
  • Nanobodies of the invention as defined herein
  • polypeptides of the invention are much preferred, it will be clear that on the basis of the description herein, the skilled person will also be able to design and/or generate, in an analogous manner, other amino acid sequences and in particular (single) domain antibodies against a metalloproteinase from the ADAM family , as well as polypeptides comprising such (single) domain antibodies.
  • Suitable scaffolds and techniques for such CDR grafting will be clear to the skilled person and are well known in the art, see for example US- A-7,180,370, WO 01/27160, EP 0 605 522, EP 0 460 167, US-A-7,054,297, Nicaise et al, Protein Science (2004), 13:1882-1891; Ewert et al., Methods, 2004 Oct; 34(2): 184- 199; Kettleborough et al., Protein Eng. 1991 Oct; 4(7): 773-783; O'Brien and Jones, Methods MoI. Biol. 2003: 207: 81-100; Skerra, J. MoI. Recognit.
  • Nanobodies of the inventions contain one or more other CDR sequences than the preferred CDR sequences mentioned above, these CDR sequences can be obtained in any manner known per se, for example from Nanobodies (preferred), VH domains from conventional antibodies (and in particular from human antibodies), heavy chain antibodies, conventional 4-chain antibodies (such as conventional human 4-chain antibodies) or other immunoglobulin sequences directed against a metalloproteinase from the ADAM family.
  • Such immunoglobulin sequences directed against a metalloproteinase from the ADAM family can be generated in any manner known per se, as will be clear to the skilled person, i.e.
  • Suitable techniques for generating such immunoglobulin sequences will be clear to the skilled person, and for example include the screening techniques reviewed by Hoogenboom, Nature Biotechnology, 23, 9, 1105-1116 (2005)
  • Other techniques for generating immunoglobulins against a specified target include for example the Nanoclone technology (as for example described in the published US patent application 2006-0211088), so-called SLAM technology (as for example described in the European patent application 0 542 810), the use of transgenic mice expressing human immunoglobulins or the well-known hybridoma techniques (see for example Larrick et al, Biotechnology, Vol.7, 1989, p. 934).
  • the CDR' s of such immunoglobulins can be used in the Nanobodies of the invention, i.e. as outlined above.
  • the sequence of such a CDR can be determined, synthesized and/or isolated, and inserted into the sequence of aNanobody of the invention (e.g. so as to replace the corresponding native CDR), all using techniques known per se such as those described herein, or Nanobodies of the invention containing such CDR's (or nucleic acids encoding the same) can be synthesized de novo, again using the techniques mentioned herein.
  • amino acid sequences of the invention can be linked to a suitable carrier or solid support so as to provide a medium than can be used in a manner known per se to purify a metalloproteinase from the ADAM family from compositions and preparations comprising the same.
  • Derivatives of the amino acid sequences of the invention that comprise a suitable detectable label can also be used as markers to determine (qualitatively or quantitatively) the presence of a metalloproteinase from the ADAM family in a composition or preparation or as a marker to selectively detect the presence of a metalloproteinase from the ADAM family on the surface of a cell or tissue (for example, in combination with suitable cell sorting techniques).
  • FIG. 3 is a similar graph to Figure 2, showing the effect of increased Nanobody concentration on ADAM 17 activity.
  • FIG. 5 is a graph showing Effect of the increasing amount of purified nanobodies (excess relative to ADAMlO) on the activity of recombinant ADAMlO (R&D system).
  • FIG. 6 is a graph showing the effect of 50 molar excess of purified nanobodies on the activity of recombinant ADAMlO (R&D system). On top of each column is indicated the percent residual activity.
  • Two llamas were immunized according to standard protocols with 6 boosts of R&D Systems Cat # 2717-PG and a cocktail of ADAM (recombinant human ADAM8 (amino residues 1-497, R&D Systems, Cat. No.l031-AD, Lot No. MWP04607A), recombinant human ADAM9 (amino residues 1-697, R&D Systems, Cat. No. 939-AD, Lot No. FQO03607A), recombinant human ADAMlO (amino residues 18-672, Cat. No 936-AD, Lot No: FDW12607A), recombinant human ADAM17/TACE (amino residues 1-671, Cat.
  • ADAM8 amino residues 1-497, R&D Systems, Cat. No.l031-AD, Lot No. MWP04607A
  • recombinant human ADAM9 amino residues 1-697, R&D Systems, Cat. No. 939-AD
  • Peripheral blood mononuclear cells were prepared from blood samples using Ficoll- Hypaque according to the manufacturer's instructions. Next, total RNA extracted was extracted from these cells and used as starting material for RT-PCR to amplify Nanobody encoding gene fragments. These fragments were cloned into phagemid vector pAX50. Phage was prepared according to standard methods (see for example the prior art and applications filed by applicant cited herein) and stored at 4 0 C for further use.
  • Example 3 Selections
  • Phage libraries 131 and 132 were used for selections on the ADAMs that were used for immunization.
  • the ADAMs were coated individually at 5 ug/ml, 0.5 ug/ml or Oug/ml (control) on Nunc Maxisorp ELISA plates. Binding phages were eluted from the coated ADAMs with Trypsine (Rl).
  • Nanobody encoding fragments from Rl and R2 were recloned into the plasmid pAX51 according to standard methods and individual colonies were picked up and grown in 96 deep well plates (1 ml volume) and induced by adding IPTG for Nanobody expression.
  • Periplasmic extracts volume: ⁇ 80 ul were prepared according to standard methods (see for example the prior art and applications filed by applicant cited herein).
  • the clones were tested in an ELISA assay.
  • lug/ml of indicated ADAM were immobilized directly on Maxisorp microtiter plates (Nunc). Free binding sites were blocked using 4% Marvel in PBS.
  • 5 ul of periplasmic extract containing Nanobody of the different clones in 100 ul 2% Marvel PBST were allowed to bind to the immobilized antigen.
  • Nanobody binding was revealed using a mouse-anti-myc secondary antibody, which was after a wash step detected with a HRP-conjugated goat-anti-mouse antibody.
  • Binding specificity was determined based on OD values compared to controls having received no Nanobody (low control, figure 1 for Rl and figure 2 for R2) Table B-I. Positive clones identify by ELISA for each ADAM after the first round of selection (left table) and second round of selection (right table). Are depicted the number of positive identified as well as the representative percentage of positive.
  • nanobodies 30F2, 30Dl, 30B6 also 30E12, but not shown here
  • Nanobodies 41B5, 30E6, 30Cl, 31E12 and 30G12 have an activating effect.
  • the activity of amino acid sequences or polypeptides of the invention on ADAM-TS5 can be tested using an commercially available ELISA-based kit (from Invitek).
  • ELISA-based kit from Invitek
  • cleavage of CD44 into the medium of cell can be used to detect the activity of ADAMlO in U251 glioblastoma (S. Atkinson and G. Murphy, publication submitted).
  • HB-EGF conjugated Alkaline phosphatase For testing the activity of of amino acid sequences or polypeptides of the invention on ADAM17 in a cell-based assay, the release of HB-EGF conjugated Alkaline phosphatase from transfected MCF7 cells can be used a readout for ADAM 17 activity (S. Atkinson and G. Murphy, submitted).
  • cleavage of CD44 into the medium of cell can be used to detect the activity of ADAMlO in U251 glioblastoma (S. Atkinson and G. Murphy, publication submitted).
  • ADAM 17 in a cell-based assay the release of HB-EGF conjugated Alkaline phosphatase from transfected MCF7 cells can be used a readout for ADAM 17 activity (S. Atkinson and G. Murphy, submitted).
  • Table B-2 gives the amino acid sequences of a number of Nanobodies against some representative ADAM proteinases.
  • Table B-2 Nanobodies against some representative ADAM metalloproteinases.
  • SEQ ID NO's 868 to 886 Nanobodies against ADAM8
  • SEQ ID NO's 908 to 931 Nanobodies against ADAMlO
  • SEQ ID NO's 932 to 948 and 1048 to 1052 Nanobodies against ADAM17
EP07857865A 2006-12-19 2007-12-19 Gegen metalloproteinase aus der adam-familie gerichtete aminosäuresequenzen und diese enthaltende polypeptide zur behandlung von mit adam in zusammenhang stehenden krankheiten und störungen Ceased EP2102244A2 (de)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP12164303A EP2514767A1 (de) 2006-12-19 2007-12-19 Gegen Metalloproteinase der ADAM-Familie gerichtete Aminosäuren und Polypeptide damit zur Behandlung von Erkrankungen und Störungen im Zusammenhang mit ADAM

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US87583406P 2006-12-19 2006-12-19
PCT/EP2007/064244 WO2008074840A2 (en) 2006-12-19 2007-12-19 Amino acid sequences directed against a metalloproteinase from the adam family and polypeptides comprising the same for the treatment of adam-related diseases and disorders

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP12164303A Division EP2514767A1 (de) 2006-12-19 2007-12-19 Gegen Metalloproteinase der ADAM-Familie gerichtete Aminosäuren und Polypeptide damit zur Behandlung von Erkrankungen und Störungen im Zusammenhang mit ADAM

Publications (1)

Publication Number Publication Date
EP2102244A2 true EP2102244A2 (de) 2009-09-23

Family

ID=39536786

Family Applications (2)

Application Number Title Priority Date Filing Date
EP12164303A Withdrawn EP2514767A1 (de) 2006-12-19 2007-12-19 Gegen Metalloproteinase der ADAM-Familie gerichtete Aminosäuren und Polypeptide damit zur Behandlung von Erkrankungen und Störungen im Zusammenhang mit ADAM
EP07857865A Ceased EP2102244A2 (de) 2006-12-19 2007-12-19 Gegen metalloproteinase aus der adam-familie gerichtete aminosäuresequenzen und diese enthaltende polypeptide zur behandlung von mit adam in zusammenhang stehenden krankheiten und störungen

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP12164303A Withdrawn EP2514767A1 (de) 2006-12-19 2007-12-19 Gegen Metalloproteinase der ADAM-Familie gerichtete Aminosäuren und Polypeptide damit zur Behandlung von Erkrankungen und Störungen im Zusammenhang mit ADAM

Country Status (5)

Country Link
US (1) US9156914B2 (de)
EP (2) EP2514767A1 (de)
AU (1) AU2007336243B2 (de)
CA (1) CA2672965C (de)
WO (1) WO2008074840A2 (de)

Families Citing this family (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008020079A1 (en) 2006-08-18 2008-02-21 Ablynx N.V. Amino acid sequences directed against il-6r and polypeptides comprising the same for the treatment of deseases and disorders associated with il-6-mediated signalling
US8444976B2 (en) 2008-07-02 2013-05-21 Argen-X B.V. Antigen binding polypeptides
JP2010195733A (ja) * 2009-02-26 2010-09-09 Rohto Pharmaceut Co Ltd 新規ペプチド
US9265834B2 (en) 2009-03-05 2016-02-23 Ablynx N.V. Stable formulations of polypeptides and uses thereof
US10005830B2 (en) 2009-03-05 2018-06-26 Ablynx N.V. Antigen binding dimer-complexes, methods of making/avoiding and uses thereof
WO2010115995A2 (en) * 2009-04-10 2010-10-14 Ablynx Nv Improved amino acid sequences directed against il-6r and polypeptides comprising the same for the treatment of il-6r related diseases and disorders
JP5647222B2 (ja) 2009-04-10 2014-12-24 アブリンクス エン.ヴェー. Il−6r関連疾患及び障害の治療のためのil−6rに指向性を有する改善されたアミノ酸配列及びこれを含むポリペプチド
US9884117B2 (en) 2009-09-03 2018-02-06 Ablynx N.V. Stable formulations of polypeptides and uses thereof
PL2533761T3 (pl) 2010-02-11 2019-09-30 Ablynx N.V. Sposoby i kompozycje do wytwarzania aerozoli
MX349886B (es) 2011-06-10 2017-08-17 Medimmune Llc Moleculas de union anti-pseudomonas lectina pisum satinum (psl) y usos de las mismas.
WO2013041722A1 (en) 2011-09-23 2013-03-28 Ablynx Nv Prolonged inhibition of interleukin-6 mediated signaling
PT2776065T (pt) * 2011-11-07 2020-09-25 Medimmune Ltd Terapias de combinação que usam moléculas de ligação de psl e pcrv anti-pseudomonas
KR101421629B1 (ko) 2012-03-12 2014-07-22 아이진 주식회사 동맥경화 예방 또는 치료용 약제학적 조성물
JP6605957B2 (ja) 2013-01-04 2019-11-13 シトムクス セラピューティクス,インコーポレイティド 生体系におけるプロテアーゼ活性を検出するための組成物及び方法
WO2016059624A2 (en) * 2014-10-15 2016-04-21 Prendergast Patrick T Compositions and methods for treatment of diseases
EP3797786A1 (de) * 2014-10-15 2021-03-31 Byrock Technologies Limited Zusammensetzungen und verfahren zur behandlung von erkrankungen
WO2016062766A1 (en) 2014-10-21 2016-04-28 Ablynx Nv Treatment of il-6r related diseases
EP3558368A4 (de) * 2016-12-23 2020-12-30 MacroGenics, Inc. Adam9-bindende moleküle und verfahren zur verwendung davon
KR20200015912A (ko) * 2017-06-02 2020-02-13 메르크 파텐트 게엠베하 Adamts5, mmp13 및 아그레칸 결합성 폴리펩타이드
AU2018277343A1 (en) * 2017-06-02 2020-01-02 Ablynx N.V. Adamts binding immunoglobulins
JP2020521804A (ja) * 2017-06-02 2020-07-27 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung Mmp13結合免疫グロブリン
CN110872354B (zh) * 2018-09-04 2022-11-01 华瑞同康生物技术(深圳)有限公司 哺乳动物细胞重组抗人tk1的鸡源的单克隆抗体、单链抗体及其制备方法和应用
JP2022520998A (ja) * 2019-02-22 2022-04-04 アンウィタ バイオサイエンシス, インク. アルブミン結合抗体及びその使用
CN114466864A (zh) * 2019-06-21 2022-05-10 索瑞索制药公司 多肽
CA3146123A1 (en) * 2019-07-29 2021-02-04 Russell B. Wilson Antibodies to candida and uses thereof
WO2021102063A1 (en) 2019-11-20 2021-05-27 Anwita Biosciences, Inc. Cytokine fusion proteins, and their pharmaceutical compositions and therapeutic applications
CN113234151B (zh) * 2021-06-08 2022-02-15 皖南医学院第一附属医院(皖南医学院弋矶山医院) 一种基于靶向SARS-CoV2病毒S蛋白的三聚体纳米抗体的CAR-NK的研制

Family Cites Families (113)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4559157A (en) 1983-04-21 1985-12-17 Creative Products Resource Associates, Ltd. Cosmetic applicator useful for skin moisturizing
LU84979A1 (fr) 1983-08-30 1985-04-24 Oreal Composition cosmetique ou pharmaceutique sous forme aqueuse ou anhydre dont la phase grasse contient un polyether oligomere et polyethers oligomeres nouveaux
US4820508A (en) 1987-06-23 1989-04-11 Neutrogena Corporation Skin protective composition
US4992478A (en) 1988-04-04 1991-02-12 Warner-Lambert Company Antiinflammatory skin moisturizing composition and method of preparing same
US4938949A (en) 1988-09-12 1990-07-03 University Of New York Treatment of damaged bone marrow and dosage units therefor
JP2919890B2 (ja) 1988-11-11 1999-07-19 メディカル リサーチ カウンスル 単一ドメインリガンド、そのリガンドからなる受容体、その製造方法、ならびにそのリガンドおよび受容体の使用
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
SE509359C2 (sv) 1989-08-01 1999-01-18 Cemu Bioteknik Ab Användning av stabiliserade protein- eller peptidkonjugat för framställning av ett läkemedel
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
WO1992002551A1 (en) 1990-08-02 1992-02-20 B.R. Centre Limited Methods for the production of proteins with a desired function
DK0605522T3 (da) 1991-09-23 2000-01-17 Medical Res Council Fremgangsmåde til fremstilling af humaniserede antistoffer
ATE165113T1 (de) 1992-05-08 1998-05-15 Creative Biomolecules Inc Mehrwertige chimäre proteine anologe und verfahren zu deren anwendungen
WO1994002610A1 (en) 1992-07-17 1994-02-03 Dana-Farber Cancer Institute Method of intracellular binding of target molecules
PT1498427E (pt) 1992-08-21 2010-03-22 Univ Bruxelles Imunoglobulinas desprovidas de cadeias leves
US6765087B1 (en) * 1992-08-21 2004-07-20 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
DK0698097T3 (da) 1993-04-29 2001-10-08 Unilever Nv Produktion af antistoffer eller (funktionaliserede) fragmenter deraf afledt af Camelidae-immunoglobuliner med tung kæde
US5910488A (en) 1993-06-07 1999-06-08 Vical Incorporated Plasmids suitable for gene therapy
AU7123494A (en) 1993-06-09 1995-01-03 Nederlandse Organisatie Voor Toegepast- Natuurwetenschappelijk Onderzoek Tno Process for producing fusion proteins comprising scfv fragments by a transformed mould
FR2708622B1 (fr) 1993-08-02 1997-04-18 Raymond Hamers Vecteur recombinant contenant une séquence d'un gène de lipoprotéine de structure pour l'expression de séquences de nucléotides.
US6091639A (en) 1993-08-27 2000-07-18 Kabushiki Kaisha Toshiba Non-volatile semiconductor memory device and data programming method
US6146826A (en) 1993-09-10 2000-11-14 The Trustees Of Columbia University In The City Of New York Green fluorescent protein
WO1995021191A1 (en) 1994-02-04 1995-08-10 William Ward Bioluminescent indicator based upon the expression of a gene for a modified green-fluorescent protein
AU1925195A (en) 1994-02-22 1995-09-04 Dana-Farber Cancer Institute Nucleic acid delivery system, method of synthesis and uses thereof
US5625048A (en) 1994-11-10 1997-04-29 The Regents Of The University Of California Modified green fluorescent proteins
US5585466A (en) 1994-12-06 1996-12-17 The United States Of America As Represented By The Administrator Of The National Aeronautics And Space Administration Crystals of serum albumin for use in genetic engineering and rational drug design
EP0739981A1 (de) 1995-04-25 1996-10-30 Vrije Universiteit Brussel Variable Fragmente von Immunglobulinen-Verwendung zur therapeutischen oder veterinären Zwecken
US5693492A (en) 1995-05-05 1997-12-02 Merck & Co., Inc. DNA encoding glutamate gated chloride channels
JPH11509088A (ja) 1995-06-23 1999-08-17 プレジデント アンド フェローズ オブ ハーバード カレッジ 血管内皮増殖因子受容体をコードする遺伝子の転写調節
AU4482996A (en) 1995-09-22 1997-04-09 Novo Nordisk A/S Novel variants of green fluorescent protein, gfp
US6027881A (en) 1996-05-08 2000-02-22 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Mutant Aequorea victoria fluorescent proteins having increased cellular fluorescence
CA2258518C (en) 1996-06-27 2011-11-22 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Recognition molecules interacting specifically with the active site or cleft of a target molecule
US6124128A (en) 1996-08-16 2000-09-26 The Regents Of The University Of California Long wavelength engineered fluorescent proteins
WO1998021355A1 (en) 1996-11-15 1998-05-22 Life Technologies, Inc. Mutants of green fluorescent protein
WO1998022141A2 (en) 1996-11-19 1998-05-28 Sangstat Medical Corporation Enhanced effects for hapten conjugated therapeutics
US6329516B1 (en) 1997-04-28 2001-12-11 Fmc Corporation Lepidopteran GABA-gated chloride channels
FR2766826B1 (fr) 1997-08-04 2001-05-18 Pasteur Institut Vecteurs derives d'anticorps pour le transfert de substances dans les cellules
ATE461282T1 (de) 1997-10-27 2010-04-15 Bac Ip Bv Multivalente antigenbindende proteine
BR9907241A (pt) 1998-01-26 2000-10-17 Unilever Nv Biblioteca de expressão, processo para preparar a mesma, uso de uma fonte não imunizada de sequências de ácido nucleico, e, processos para preparar fragmentos de anticorpos e, para preparar um anticorpo
KR20010034512A (ko) 1998-02-19 2001-04-25 베렌슨, 론 림프구 활성화 조절을 위한 조성물 및 그 방법
EP0967284A1 (de) 1998-05-28 1999-12-29 Pfizer Limited Phosphodiesterasen
GB9824632D0 (en) 1998-11-10 1999-01-06 Celltech Therapeutics Ltd Biological compounds
IL127127A0 (en) 1998-11-18 1999-09-22 Peptor Ltd Small functional units of antibody heavy chain variable regions
WO2000040968A1 (en) 1999-01-05 2000-07-13 Unilever Plc Binding of antibody fragments to solid supports
WO2000043507A1 (en) 1999-01-19 2000-07-27 Unilever Plc Method for producing antibody fragments
CA2361678A1 (en) 1999-02-05 2000-08-10 Rijksuniversiteit Leiden Method of modulating metabolite biosynthesis in recombinant cells
DE1100895T1 (de) 1999-03-15 2001-09-06 Univ British Columbia Abc1 polypeptide und verfahren und reagenzien zur modulation des cholesterolgehalts
CN100434441C (zh) 1999-04-22 2008-11-19 荷兰联合利华有限公司 利用单价抗原-结合蛋白抑制病毒感染
KR20020012612A (ko) 1999-06-18 2002-02-16 씨브이 쎄러퓨틱스, 인코포레이티드 Atp 결합 카세트 트랜스포터 단백질 abc1을사용하여 콜레스테롤 유출을 증가시키고 hdl을상승시키기 위한 조성물 및 방법
ES2355055T3 (es) 1999-07-28 2011-03-22 Genentech, Inc. Composiciones y procedimientos para el tratamiento de tumores.
IL147920A0 (en) 1999-08-02 2002-08-14 Keygene Nv Method for generating cgmmv resistant plants, genetic constructs, and the obtained cgmmv-resistant plants
GB9922124D0 (en) 1999-09-17 1999-11-17 Pfizer Ltd Phosphodiesterase enzymes
US6479280B1 (en) 1999-09-24 2002-11-12 Vlaams Interuniversitair Institutuut Voor Biotechnologie Vzw Recombinant phages capable of entering host cells via specific interaction with an artificial receptor
US6849425B1 (en) 1999-10-14 2005-02-01 Ixsys, Inc. Methods of optimizing antibody variable region binding affinity
DE19955408A1 (de) 1999-11-18 2001-05-23 Bayer Ag GABA-B-Rezeptoren
ATE440111T1 (de) 1999-11-29 2009-09-15 Bac Ip B V Immobilisierte antigenbindende moleküle aus einer domäne
EP1242460B1 (de) 1999-11-29 2006-10-18 Unilever Plc Immobilisierung von proteinen mit hilfe eines polypeptidsegments
CA2390691C (en) 1999-12-24 2016-05-10 Genentech, Inc. Methods and compositions for prolonging elimination half-times of bioactive compounds
ATE428733T1 (de) 2000-03-14 2009-05-15 Unilever Nv Variabele domänen der schweren kette eines antikörpers gegen menschliche ernährungslipasen und deren verwendungen
EP1142910A1 (de) 2000-04-07 2001-10-10 Jürgen Prof. Dr. Frey Hemmstoffe der Bildung von löslichem humanen CD23
CA2405912A1 (en) 2000-04-12 2001-10-18 Human Genome Sciences, Inc. Albumin fusion proteins
CA2380443C (en) 2000-05-26 2013-03-12 Ginette Dubuc Single-domain antigen-binding antibody fragments derived from llama antibodies
US7943129B2 (en) 2000-05-26 2011-05-17 National Research Council Of Canada Single-domain brain-targeting antibody fragments derived from llama antibodies
US6741957B1 (en) 2000-07-21 2004-05-25 Daimlerchrysler Corporation Analytical tire model for vehicle durability and ride comfort analysis
WO2002048193A2 (en) 2000-12-13 2002-06-20 Unilever N.V. Camelidae antibody arrays
US7054297B1 (en) 2000-12-28 2006-05-30 Cisco Technology, Inc. Distribution of packets to high data rate communications devices using multicast protocols
US7829084B2 (en) 2001-01-17 2010-11-09 Trubion Pharmaceuticals, Inc. Binding constructs and methods for use thereof
RU2420537C2 (ru) 2001-01-17 2011-06-10 Трабьон Фармасьютикалз Инк. Слитые белки связывающий домен-иммуноглобулин
JP4336771B2 (ja) 2001-03-09 2009-09-30 モルフォシス アーゲー 血清アルブミン結合部分
GB0110029D0 (en) 2001-04-24 2001-06-13 Grosveld Frank Transgenic animal
US20060073141A1 (en) 2001-06-28 2006-04-06 Domantis Limited Compositions and methods for treating inflammatory disorders
AU2002319402B2 (en) 2001-06-28 2008-09-11 Domantis Limited Dual-specific ligand and its use
WO2003014960A2 (en) 2001-08-03 2003-02-20 Medical Research Council Method of identifying a consensus sequence for intracellular antibodies
US7371849B2 (en) 2001-09-13 2008-05-13 Institute For Antibodies Co., Ltd. Methods of constructing camel antibody libraries
FR2829940A1 (fr) 2001-09-27 2003-03-28 Synt Em Compositions pour la vectorisation d'anticorps a travers la barriere hematoencephalique et leur utilisation pour le diagnostic ou le traitement des maladies du systeme nerveux central
JP2005289809A (ja) 2001-10-24 2005-10-20 Vlaams Interuniversitair Inst Voor Biotechnologie Vzw (Vib Vzw) 突然変異重鎖抗体
KR100599789B1 (ko) 2001-12-03 2006-07-12 삼성에스디아이 주식회사 방열효율이 향상된 플라즈마 디스플레이 장치 및 그 제조방법
AU2002351896A1 (en) 2001-12-11 2003-06-23 Ablynx N.V. Method for displaying loops from immunoglobulin domains in different contexts
US20050037358A1 (en) 2001-12-21 2005-02-17 Serge Muyldermans Method for cloning of variable domain sequences
CA2471645A1 (en) 2002-01-03 2003-07-10 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Immunoconjugates useful for treatment of tumours
WO2003066822A2 (en) * 2002-02-05 2003-08-14 Wyeth Truncated aggrecanase molecules
US20060002935A1 (en) 2002-06-28 2006-01-05 Domantis Limited Tumor Necrosis Factor Receptor 1 antagonists and methods of use therefor
JP2006512895A (ja) 2002-06-28 2006-04-20 ドマンティス リミテッド リガンド
US7004940B2 (en) 2002-10-10 2006-02-28 Ethicon, Inc. Devices for performing thermal ablation having movable ultrasound transducers
JP2006512910A (ja) 2002-10-23 2006-04-20 ルードヴィッヒ インスティテュート フォー キャンサー リサーチ A34およびa33様3dna、タンパク質、それらに対する抗体、ならびに同一物を使用した治療方法
EP1900753B1 (de) 2002-11-08 2017-08-09 Ablynx N.V. Verfahren zur Verabreichung therapeutischer Polypeptide und Polypeptide dafür
US20060228355A1 (en) 2003-11-07 2006-10-12 Toon Laeremans Camelidae single domain antibodies vhh directed against epidermal growth factor receptor and uses therefor
EP2267032A3 (de) 2002-11-08 2011-11-09 Ablynx N.V. Verfahren zur Verabreichung therapeutischer Polypeptide und Polypeptide dafür
GB0228210D0 (en) 2002-12-03 2003-01-08 Babraham Inst Single chain antibodies
ES2374068T3 (es) 2002-12-03 2012-02-13 Ucb Pharma, S.A. Ensayo para identificar células productoras de anticuerpos.
GB0230203D0 (en) 2002-12-27 2003-02-05 Domantis Ltd Fc fusion
CA2509153C (en) 2002-12-31 2013-04-16 Nektar Therapeutics Al, Corporation Hydrolytically stable maleimide-terminated polymers
CA2512545C (en) 2003-01-10 2015-06-30 Karen Silence Recombinant vhh single domain antibody from camelidae against von willebrand factor (vwf)
GB0312481D0 (en) 2003-05-30 2003-07-09 Celltech R&D Ltd Antibodies
EP1639465B1 (de) 2003-06-30 2011-04-06 Continental Automotive GmbH Verfahren zur überwachung des programmlaufs in einem mikro-computer
PL1639011T3 (pl) 2003-06-30 2009-05-29 Domantis Ltd Pegilowane przeciwciała jednodomenowe (dAb)
JP2007521234A (ja) 2003-08-12 2007-08-02 ウィリアム エム ヤーブロー 尋常性ざ瘡の治療薬及び使用方法
EP1512696A1 (de) 2003-08-14 2005-03-09 Diatos Aminsäuresequenz, die den Transfer von Substanzen von Interesse in das Innere von Zellen und/oder Zellkernen erleichtern
WO2005019824A1 (en) 2003-08-20 2005-03-03 Celltech R & D Limited Methods for obtaining antibodies
AU2003304638A1 (en) * 2003-12-04 2005-07-14 Wyeth Biaryl sulfonamides and methods for using same
US7207410B2 (en) 2004-04-29 2007-04-24 Daimlerchrysler Corporation Apparatus and method for enhanced impact sensing
US20050279676A1 (en) 2004-06-21 2005-12-22 Izzy Zuhair A Fluid filter assembly for a dispensing faucet
US20060008601A1 (en) 2004-06-25 2006-01-12 Zeik Douglas B Flexible laminate having an integrated pressure release valve
KR101151957B1 (ko) 2004-07-22 2012-06-01 로저 킹돈 크레이그 결합 분자
US7180370B2 (en) 2004-09-01 2007-02-20 Micron Technology, Inc. CMOS amplifiers with frequency compensating capacitors
US7563443B2 (en) 2004-09-17 2009-07-21 Domantis Limited Monovalent anti-CD40L antibody polypeptides and compositions thereof
EP1814917A2 (de) 2004-10-13 2007-08-08 Ablynx N.V. Anti-amyloid-beta-antikörper aus kameliden und diese enthaltende polypeptide für die behandlung und diagnose degenerativer nervenerkrankungen wie etwa morbus alzheimer
WO2006040154A2 (en) 2004-10-14 2006-04-20 Dublin City University Prokaryotic two hybrid system
JP5616572B2 (ja) * 2004-12-17 2014-10-29 モナッシュ ユニバーシティ 哺乳類修飾adam10プロテアーゼまたはそのフラグメント、単離核酸、遺伝子構築物、宿主細胞、抗体、及び産生方法
JP2008528010A (ja) 2005-01-31 2008-07-31 アブリンクス ナームローゼ フェンノートシャップ 重鎖抗体の可変ドメイン配列を作出する方法
AU2006210589B2 (en) * 2005-02-02 2011-12-08 Macrogenics West, Inc. ADAM-9 modulators
EP2949668B1 (de) * 2005-05-18 2019-08-14 Ablynx N.V. Verbesserte nanokörper-tm gegen tumornekrosefaktor alpha
CA2608873C (en) 2005-05-20 2017-04-25 Ablynx Nv Single domain vhh antibodies against von willebrand factor

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
DEFFAR KHALISSA ET AL: "Nanobodies - the new concept in antibody engineering", AFRICAN JOURNAL OF BIOTECHNOLOGY, ACADEMIC PRESS, US, vol. 8, no. 12, 17 June 2009 (2009-06-17), pages 2645 - 2652, XP009136924, ISSN: 1684-5315 *
GIBBS W W: "NANOBODIES", SCIENTIFIC AMERICAN, SCIENTIFIC AMERICAN INC., NEW YORK, NY, US, vol. 293, no. 2, 1 August 2005 (2005-08-01), pages 67 - 71, XP001238880, ISSN: 0036-8733 *
GLASSON S S ET AL: "Deletion of active ADAMTS5 prevents cartilage degradation in a murine model of osteoarthritis", NATURE, NATURE PUBLISHING GROUP, LONDON, GB, vol. 434, 31 March 2005 (2005-03-31), pages 644 - 648, XP003003284, ISSN: 0028-0836, DOI: DOI:10.1038/NATURE03369 *
HOLLIGER PHILIPP ET AL: "ENGINEERED ANTIBODY FRAGMENTS AND THE RISE OF SINGLE DOMAINS", NATURE BIOTECHNOLOGY, NATURE PUBLISHING GROUP, NEW YORK, NY, US, vol. 23, no. 9, 1 September 2005 (2005-09-01), pages 1126 - 1136, XP008076746, ISSN: 1087-0156, DOI: 10.1038/NBT1142 *
MITSUTOSHI NAKADA ET AL: "Human glioblastomas overexpress ADAMTS-5 that degrades brevican", ACTA NEUROPATHOLOGICA, SPRINGER, BERLIN, DE, vol. 110, no. 3, 1 September 2005 (2005-09-01), pages 239 - 246, XP019340780, ISSN: 1432-0533, DOI: DOI:10.1007/S00401-005-1032-6 *
REVETS H ET AL: "NANOBODIES AS NOVEL AGENTS FOR CANCER THERAPY", EXPERT OPINION ON BIOLOGICAL THERAPY, ASHLEY, LONDON, GB, vol. 5, no. 1, 1 January 2005 (2005-01-01), pages 111 - 124, XP009076361, ISSN: 1471-2598, DOI: 10.1517/14712598.5.1.111 *
STANTON H ET AL: "ADAMTS5 is the major aggrecanase in mouse cartilage in vivo and in vitro", 20050101, vol. 434, 1 January 2005 (2005-01-01), pages 648 - 652, XP003003285 *
VASILII V KLIMOV: "Spontaneous emission of an atom in the presence of nanobodies", QUANTUM ELECTRONICS, TURPION LTD., LONDON, GB, vol. 31, no. 7, 1 January 2001 (2001-01-01), pages 569 - 586, XP009160370, ISSN: 1063-7818, DOI: 10.1070/QE2001V031N07ABEH002007 *
VOROS G ET AL: "Expression of aggrecan(ases) during murine preadipocyte differentiation and adipose tissue development", BIOCHIMICA ET BIOPHYSICA ACTA - GENERAL SUBJECTS, ELSEVIER SCIENCE PUBLISHERS, NL, vol. 1760, no. 12, 1 December 2006 (2006-12-01), pages 1837 - 1844, XP025014896, ISSN: 0304-4165, [retrieved on 20061201], DOI: DOI:10.1016/J.BBAGEN.2006.08.016 *

Also Published As

Publication number Publication date
WO2008074840A3 (en) 2008-10-30
EP2514767A1 (de) 2012-10-24
AU2007336243A1 (en) 2008-06-26
CA2672965A1 (en) 2008-06-26
US20100150939A1 (en) 2010-06-17
CA2672965C (en) 2018-02-06
WO2008074840A2 (en) 2008-06-26
AU2007336243B2 (en) 2012-07-26
US9156914B2 (en) 2015-10-13

Similar Documents

Publication Publication Date Title
US9156914B2 (en) Amino acid sequences directed against a metalloproteinase from the ADAM family and polypeptides comprising the same for the treatment of ADAM-related diseases and disorders
US11078290B2 (en) Amino acid sequences directed against RANK-L and polypeptides comprising the same for the treatment of bone diseases and disorders
US8557965B2 (en) Single variable domains against notch pathway members
AU2016204023B2 (en) Amino acid sequences directed against the angiopoietin/tie system and polypeptides comprising the same for the treatment of diseases and disorders related to angiogenesis
US20100136018A1 (en) Anti-FC-receptor single domain antibodies (nanobodies-tm) and therapeutic use
US20090297535A1 (en) Amino Acid Sequences Directed Against Il-6 And Polypetides Comprising The Same For The Treatment Of Diseases And Disorders Associated With Il-6 Mediated Signalling
US20110118185A9 (en) Amino acid sequences directed against vascular endothelial growth factor and polypeptides comprising the same for the treatment of conditions and diseases characterized by excessive and/or pathological angiogenesis or neovascularization
AU2012201229B2 (en) Amino acid sequences directed against Rank-L and polypeptides comprising the same for the treatment of bone diseases and disorders

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20090707

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20100406

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: ABLYNX N.V.

APBK Appeal reference recorded

Free format text: ORIGINAL CODE: EPIDOSNREFNE

APBN Date of receipt of notice of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA2E

APBR Date of receipt of statement of grounds of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA3E

APAF Appeal reference modified

Free format text: ORIGINAL CODE: EPIDOSCREFNE

APBT Appeal procedure closed

Free format text: ORIGINAL CODE: EPIDOSNNOA9E

REG Reference to a national code

Ref country code: DE

Ref legal event code: R003

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN REFUSED

18R Application refused

Effective date: 20160912